1
|
Zhang B, He C, Su T, Qin Z, Cheng Q, Zhu Z, Chen M, Yu W. Association and dynamic change of fecal calprotectin with sepsis associated liver dysfunction in adults with sepsis. Sci Rep 2025; 15:16192. [PMID: 40346167 PMCID: PMC12064738 DOI: 10.1038/s41598-025-98904-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Fecal calprotectin (FC) is a biomarker of gut inflammation but its association with sepsis associated liver dysfunction (SALD) is unclear. This single-center, prospective cohort study investigated the relationship between FC and SALD in adults with sepsis. FC concentrations were measured on days 1 (FC1) and 3 (FC3). The difference between FC3 and FC1 (∆FC) was calculated. The relationship between FC and SALD was assessed using multivariate analysis. Receiver operating characteristic curves were used to assess the predictive value of SALD biomarkers. Ninety-five patients with sepsis (33 with SALD and 62 without SALD) were enrolled between January 2023 and March 2024. The median FC3 level and ∆FC were significantly higher in the SALD group than in the non-SALD group (both p < 0.001), whereas the median FC1 level did not differ significantly between groups. FC3 level and ∆FC were positively associated with SALD in the adjusted analysis (both p < 0.001) with areas under the curve of 0.784 and 0.848, respectively, and cut-off values of 930.88 μg/g and - 8.80 μg/g, respectively. In adults with sepsis, SALD occurrence was more strongly associated with elevated FC3 levels and ∆FC than with FC1 levels; therefore, monitoring FC3 levels and ∆FC could help detect SALD.
Collapse
Affiliation(s)
- Beiyuan Zhang
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Chenzhe He
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Ting Su
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Zimeng Qin
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Qi Cheng
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Zhanghua Zhu
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Ming Chen
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China.
| | - Wenkui Yu
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China.
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China.
| |
Collapse
|
2
|
Sthity RA, Islam MZ, Sagar MEK, Gazi MA, Ferdous J, Kabir MM, Mahfuz M, Ahmed T, Mostafa I. Association of Escherichia coli pathotypes with fecal markers of enteropathy and nutritional status among underweight adults in Bangladesh. Front Cell Infect Microbiol 2025; 15:1553688. [PMID: 40276386 PMCID: PMC12018316 DOI: 10.3389/fcimb.2025.1553688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Environmental enteric dysfunction (EED), a subclinical intestinal disorder, is characterized by chronic fecal-oral exposure to entero-pathogens and could be diagnosed by measuring non-invasive biomarkers. Escherichia coli is the one of the key bacterial enteric pathogens that drives EED, but there is a lack of information on the E. coli pathotypes in relation to the biomarkers of EED in malnourished adults. Here, we intended to measure the possible association of these pathotypes with EED biomarkers and nutritional status of adults residing in a slum in Bangladesh. Method Fecal samples were collected from 524 malnourished adults (BMI ≤18.5 kg/m2) living in a slum-setting in Dhaka from March 2016 to September 2019 and analyzed by TaqMan Array Card assays to evaluate the presence of E. coli pathotypes and other entero-pathogens. The multivariable linear regression model was used to assess the association. Results In these malnourished adults, the most prevalent pathotype of E. coli was EAEC (61.7%) and the least prevalent was STEC (6.7%). The prevalence of atypical EPEC, ETEC and Shigella/EIEC were 52%, 48.9% and 45.1% respectively. The infection with atypical EPEC had significant positive association with levels of Myeloperoxidase (b = 0.38; 95% CI = 0.11, 0.65; p-value = 0.006). Similarly, a significantly higher concentration of alpha-1-antitrypsin (b = 0.13; 95% CI = 0.03, 0.22; p-value = 0.011) was found in the STEC-infected adults. However, no notable association was found between the E. coli pathotypes and nutritional status of these adult participants. Moreover, Plesiomonas infected adults were more likely to be infected with EAEC (p-value = 0.017), ETEC (p-value <0.001) and STEC (pvalue = 0.002). Significant coinfection was also detected among the pathotypes and other entero-pathogens such as Giardia, Ascaris, Campylobacter, Salmonella, Enterocytozoon bieneusi, and Adenovirus. Discussion The study results imply that there is an influence of particular E. coli pathotypes (EPEC and STEC) on intestinal inflammation and gut permeability of the malnourished Bangladeshi adults, but no association with nutritional status is found. Potential pathogenicity of the E. coli pathotypes is also observed when co-infection with other pathogens exists in these adults.
Collapse
Affiliation(s)
- Rahvia Alam Sthity
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Zahidul Islam
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Ehsanul Kabir Sagar
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Jafrin Ferdous
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mamun Kabir
- Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Office of the Executive Director, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| | - Ishita Mostafa
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
3
|
Hoermann H, Franzel J, Tautz J, Oommen PT, Lainka E, Mayatepek E, Hoehn T. Intrauterine intestinal obstruction in a preterm infant with severe mevalonate kinase deficiency - a case report. Matern Health Neonatol Perinatol 2025; 11:8. [PMID: 40038796 DOI: 10.1186/s40748-025-00207-w] [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] [Received: 10/02/2024] [Accepted: 02/09/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Mevalonate kinase deficiency is an inherited autoinflammatory disorder that can present with a wide clinical spectrum, ranging from mild forms with recurrent episodes of fever, lymphadenopathy, splenomegaly and skin rash to the much rarer severe form, which is characterized by additional occurrences of psychomotor impairment, cholestatic jaundice, ophthalmological symptoms, and failure to thrive. The few cases described with perinatal onset often showed a very severe clinical course. CASE PRESENTATION Here, we report the case of a preterm infant born at 30 + 2 weeks of gestation with a prenatal genetic diagnosis of mevalonate kinase deficiency presenting with intrauterine bowel dilatation, mild hydrops fetalis, and microcephaly. Laparotomy on the second day of life revealed intestinal obstruction necessitating partial ileum resection and ileostomy. The neonate had recurrent inflammatory reactions with elevated C-reactive protein levels, severe cholestasis, a progressive liver dysfunction, and an increasingly distended abdomen with subsequent respiratory insufficiency. Urinary mevalonic acid was highly elevated. The patient received anti-inflammatory therapy with prednisone and anakinra. Unfortunately, the patient died at the age of 77 days due to cardiorespiratory failure. CONCLUSIONS This case shows that intestinal obstruction with dilated fetal bowel loops can be an initially leading clinical symptom of severe mevalonate kinase deficiency. Diagnostics should be considered at an early stage, especially in the presence of other anomalies such as hydrops fetalis, growth restriction, or microcephaly. Data on the neonatal course of severe mevalonate kinase deficiency are still scarce and further studies are needed, particularly on treatment in neonates and young infants.
Collapse
Affiliation(s)
- Henrike Hoermann
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich Heine University, Moorenstraße 5, Duesseldorf, 40225, Germany.
| | - Julia Franzel
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich Heine University, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Juliane Tautz
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich Heine University, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Prasad T Oommen
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Medical Faculty, Division of Paediatric Rheumatology, University Hospital Duesseldorf, Heinrich Heine University, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Elke Lainka
- Children's Hospital, Department of Paediatric Gastroenterology, Hepatology, and Transplant Medicine, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany
| | - Ertan Mayatepek
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich Heine University, Moorenstraße 5, Duesseldorf, 40225, Germany
| | - Thomas Hoehn
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich Heine University, Moorenstraße 5, Duesseldorf, 40225, Germany
| |
Collapse
|
4
|
Kijmassuwan T, Balouch F. Approach to Congenital Diarrhea and Enteropathies (CODEs). Indian J Pediatr 2024; 91:598-605. [PMID: 38105403 DOI: 10.1007/s12098-023-04929-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 11/01/2023] [Indexed: 12/19/2023]
Abstract
Congenital diarrhea and enteropathies (CODEs) constitute a group of rare genetic disorders characterized by severe diarrhea and malabsorption in the neonatal period or early infancy. Timely diagnosis and treatment is essential to prevent life-threatening complications, including dehydration, electrolyte imbalance, and malnutrition. This review offers a simplified approach to the diagnosis of CODEs, with a specific focus on microvillus inclusion disease (MVID), congenital tufting enteropathy (CTE), congenital chloride diarrhea (CLD), and congenital sodium diarrhea (CSD). Patients with CODEs typically present with severe watery or occasionally bloody diarrhea, steatorrhea, dehydration, poor growth, and developmental delay. Therefore, it is crucial to thoroughly evaluate infants with diarrhea to rule out infectious, allergic, or anatomical causes before considering CODEs as the underlying etiology. Diagnostic investigations for CODEs encompass various modalities, including stool tests, blood tests, immunological studies, endoscopy and biopsies for histology and electron microscopy, and next-generation sequencing (NGS). NGS plays a pivotal role in identifying the genetic mutations responsible for CODEs. Treatment options for CODEs are limited, often relying on total parenteral nutrition for hydration and nutritional support. In severe cases, intestinal transplantation may be considered. The long-term prognosis varies among specific CODEs, with some patients experiencing ongoing intestinal failure and associated complications. In conclusion, the early recognition and accurate diagnosis of CODEs are of paramount importance for implementing appropriate management strategies. Further research and advancements in genetic testing hold promise for enhancing diagnostic accuracy and exploring potential targeted therapies for these rare genetic disorders.
Collapse
Affiliation(s)
- Teera Kijmassuwan
- Division of Gastroenterology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Gastroenterology, Queensland Children's Hospital, South Brisbane, QLD, Australia
| | - Fariha Balouch
- Department of Gastroenterology, Queensland Children's Hospital, South Brisbane, QLD, Australia.
| |
Collapse
|
5
|
Farghaly MAA, Ali MAM, Ramey S, Said W, Abdelkarem A, Collin M. Characteristics of fecal calprotectin as an early marker for suspected necrotizing enterocolitis in newborns exclusively fed maternal breast milk: a case-control study. Proc AMIA Symp 2023; 37:43-47. [PMID: 38174026 PMCID: PMC10761025 DOI: 10.1080/08998280.2023.2277580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/22/2023] [Indexed: 01/05/2024] Open
Abstract
Objective Numerous studies have proposed using fecal calprotectin among many biomarkers associated with necrotizing enterocolitis (NEC) diagnosis. This study aimed to evaluate fecal calprotectin as an early marker for suspected NEC (stage 1) in infants fed exclusively breast milk. Methods We collected 20 stool samples from newborns admitted to the neonatal intensive care unit at Aswan University Hospital diagnosed with stage I NEC. We compared them with 20 samples from matched healthy newborns. Fecal calprotectin level was measured by enzyme-linked immunosorbent assay. Results Fecal calprotectin level was higher in cases than in the control group (P < 0.001). Also, there was a positive correlation between fecal calprotectin and C-reactive protein in the studied cases (P = 0.001). However, there were no correlations between fecal calprotectin and sex or postnatal age. Conclusion Fecal calprotectin levels increase in newborns with stage I NEC. Although not specific, its sensitivity suggests a role as a potential biomarker in the evaluation of suspected NEC.
Collapse
Affiliation(s)
- Mohsen A. A. Farghaly
- Cleveland Clinic Children’s, Cleveland, Ohio, USA
- Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Mahmoud A. M. Ali
- Faculty of Medicine, Aswan University, Aswan, Egypt
- Department of Pediatrics, Case Western Reserve University/MetroHealth System, Cleveland, Ohio, USA
- Department of Pediatrics, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Stacey Ramey
- Department of Pediatrics, Case Western Reserve University/MetroHealth System, Cleveland, Ohio, USA
| | - Wafaa Said
- Faculty of Medicine, Aswan University, Aswan, Egypt
| | | | - Marc Collin
- Department of Pediatrics, Case Western Reserve University/MetroHealth System, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Kamalova AA, Garina GA, Valeeva IK, Gaifutdinova AR. Fecal calprotectin as a marker of inflammatory bowel diseases. ROSSIYSKIY VESTNIK PERINATOLOGII I PEDIATRII (RUSSIAN BULLETIN OF PERINATOLOGY AND PEDIATRICS) 2023; 68:138-143. [DOI: 10.21508/1027-4065-2023-68-5-138-143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Calprotectin is a calcium- and zinc-binding protein belonging to the S100 protein family. This protein is found mainly in the cytoplasm of neutrophils, and, to a lesser extent, in monocytes and macrophages, which can be found in any human organs, but mainly in blood, cerebrospinal fluid, feces, saliva, and synovial fluid. Calprotectin is an effective tool forthe differential diagnosis of inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS). There is a connection of fecal calprotectin (FC) with the endoscopic activity of IBD, however, the available literature shows significant differences in the sensitivity and specificity of FC for predicting the endoscopic activity of the disease. In addition, FC can be considered as a predictor of histological mucosal healing and as a marker for assessing the response to treatment, including surgical, but there is still no consensus on the threshold value of a biomarker for these purposes. Conflicting data are presented in reports on FC as a predictor of IBD recurrence. FC seems to be effective for detecting relapse, however, there is no specific threshold value, therefore, the marker cannot completely replace endoscopic examination methods. In addition, there is intraindividual variability in the concentration of FC in patients, depending on age, type of feeding in the first year of life, taking medications, which significantly complicates the interpretation of the results.
Collapse
|
7
|
Vera Chamorro JF, Sánchez Franco C, Vargas Sandoval M, Mora Quintero DV, Riveros López JP, Sarmiento Quintero F, Ortiz-Piedrahita C, Calderón-Guerrero OG, Laignelet H, Losada Gómez CL, Sánchez DP, López Panqueva RDP, Aponte Barrios W, Triana Rodríguez GA, Osorno A, Becerra Granados LM, Ortega López MC, Correa Jiménez Ó, Maradei Anaya SJ, García Acero M, Acevedo Forero AM, Prada A, Ramírez Urrego LC, Salcedo Castilla LK, Enríquez A, Suárez Fuentes MA, González Leal N, Peña Hernández S, Sotaquirá Guáqueta L, Sosa F, Fierro F, Correa S, Martín de Carpi FJ. Consenso colombiano de la enfermedad inflamatoria intestinal pediátrica. REVISTA COLOMBIANA DE GASTROENTEROLOGÍA 2023; 38:1-75. [DOI: 10.22516/25007440.943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Introducción: la colitis ulcerativa pediátrica (CUP), la enfermedad de Crohn pediátrica (ECP) y la enfermedad inflamatoria intestinal pediátrica no clasificable (EIIPNC) tienen particularidades clínicas y psicosociales que las diferencian de las del adulto y pueden condicionar enfoques terapéuticos distintos por las posibles repercusiones nutricionales, crecimiento y desarrollo, lo que representa un desafío para el pediatra y el gastroenterólogo. Objetivo: desarrollar recomendaciones basadas en la evidencia por consenso de expertos para el diagnóstico y el tratamiento oportunos y seguros de la enfermedad inflamatoria intestinal pediátrica (EIIP) en menores de 18 años, para los profesionales que atienden estos pacientes y los pagadores en salud. Metodología: a través de un panel de expertos del Colegio Colombiano de Gastroenterología, Hepatología y Nutrición Pediátrica (COLGAHNP) y un grupo multidisciplinario se formularon 35 preguntas en relación con el cuadro clínico, el diagnóstico y el tratamiento de la EIIP. A través de una revisión y un análisis crítico de la literatura, con especial énfasis en las principales guías de práctica clínica (GPC), estudios clínicos aleatorizados (ECA) y metaanálisis de los últimos 10 años, los expertos plantearon 77 recomendaciones que respondían a cada una de las preguntas de investigación con sus respectivos puntos prácticos. Posteriormente, cada una de las afirmaciones se sometieron a votación dentro del grupo desarrollador, incluyendo las afirmaciones que alcanzaron > 80 %. Resultados: todas las afirmaciones alcanzaron una votación > 80 %. La EIIP tiene mayor extensión, severidad y evolución hacia la estenosis, enfermedad perianal, manifestaciones extraintestinales y retraso en el crecimiento en comparación con los pacientes adultos, por lo que su manejo debe ser realizado por grupos multidisciplinarios liderados por gastroenterólogos pediatras y prepararlos para una transición a la edad adulta. Los criterios de Porto permiten una clasificación práctica de la EIIP. En la ECP, debemos usar la clasificación de París y debemos realizar ileocolonoscopia y esofagogastroduodenoscopia, ya que el 50 % tienen un compromiso superior, usando el SES-CD (UCEIS/Mayo en CUP) y tomando múltiples biopsias. Los laboratorios iniciales deben incluir marcadores de inflamación, calprotectina fecal y descartar infecciones intestinales. El tratamiento, la inducción y el mantenimiento de la EIIP deben ser individualizados y decididos según la estratificación de riesgo. En el seguimiento se debe usar el Pediatric Crohn Disease Activity Index (PCDAI) y Pediatric Ulcerative Colitis Activity Index (PUCAI) de las últimas 48 horas. Los pacientes con EIIP temprana e infantil, deben ser valorados por inmunólogos y genetistas. Conclusión: se proporciona una guía de consenso con recomendaciones basadas en la evidencia sobre el diagnóstico y los tratamientos oportunos y seguros en los pacientes con EIIP.
Collapse
|
8
|
Fernández-Tuñas MDC, Pérez-Muñuzuri A, Trastoy-Pena R, Pérez del Molino ML, Couce ML. Effects of Maternal Stress on Breast Milk Production and the Microbiota of Very Premature Infants. Nutrients 2023; 15:4006. [PMID: 37764789 PMCID: PMC10534677 DOI: 10.3390/nu15184006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Perinatal stress experienced by mothers of very premature newborns may influence the mother's milk and the infant's intestinal microbiota. This prospective study of mothers of very preterm infants fed with mother's own milk (MOM) was carried out in a tertiary hospital over a 2-year period. The assessment of maternal stress in 45 mothers of 52 very preterm newborns using the parental stress scale (PSS:NICU) revealed an inverse relationship between stress and MOM production in the first days of life (p = 0.012). The greatest contributor to stress was the one related to the establishment of a mother-child bond. Maternal stress was lower in mothers in whom the kangaroo method was established early (p = 0.011) and in those with a higher educational level (p = 0.032). Levels of fecal calprotectin (FC) decreased with the passage of days and were directly correlated with birthweight (p = 0.044). FC levels 7 days post-delivery were lower in newborns that received postnatal antibiotics (p = 0.027). High levels of maternal stress resulted in progressive decreases and increases in the proportions of Firmicutes and Proteobacteria species, respectively, over 15 days post-delivery, both in MOM and in fecal samples from premature newborns. These findings underscore the importance of recognizing and appropriately managing maternal stress in neonatal units, given its marked influence on both the microbiota of maternal milk and the intestinal microbiota of premature newborns.
Collapse
Affiliation(s)
- María del Carmen Fernández-Tuñas
- Department of Neonatology, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- IDIS-Health Research Institute of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS-SAMID), Carlos III Health Institute, 5 Monforte de Lemos Av., 28029 Madrid, Spain
| | - Alejandro Pérez-Muñuzuri
- Department of Neonatology, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- IDIS-Health Research Institute of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS-SAMID), Carlos III Health Institute, 5 Monforte de Lemos Av., 28029 Madrid, Spain
- Faculty of Medicine, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Rocío Trastoy-Pena
- Department of Microbiology, University Hospital of Santiago de Compostela, Santiago de Compostela University, 15706 A Coruña, Spain; (R.T.-P.); (M.L.P.d.M.)
| | - María Luisa Pérez del Molino
- Department of Microbiology, University Hospital of Santiago de Compostela, Santiago de Compostela University, 15706 A Coruña, Spain; (R.T.-P.); (M.L.P.d.M.)
| | - María L. Couce
- Department of Neonatology, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- IDIS-Health Research Institute of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin (RICORS-SAMID), Carlos III Health Institute, 5 Monforte de Lemos Av., 28029 Madrid, Spain
- Faculty of Medicine, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| |
Collapse
|
9
|
Kapel N, Ouni H, Benahmed NA, Barbot-Trystram L. Fecal Calprotectin for the Diagnosis and Management of Inflammatory Bowel Diseases. Clin Transl Gastroenterol 2023; 14:e00617. [PMID: 37440723 PMCID: PMC10522095 DOI: 10.14309/ctg.0000000000000617] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Calprotectin is a heterodimeric calcium- and zinc-binding protein mainly derived from the cytoplasm of neutrophils that has direct antimicrobial functions and a role in the regulation of the innate immune response. It can be found in various biological compartments, in particular, the stool, with concentrations related to the level of mucosal inflammation. The measurement of fecal calprotectin has thus been recognized as a useful surrogate marker to distinguish patients with inflammatory bowel disease from those with irritable bowel syndrome. Moreover, it allows the monitoring of intestinal inflammation with a high negative predictive value, making it possible to exclude the diagnosis of inflammatory bowel disease for symptomatic patients. It also shows high sensitivity for the identification of patients requiring additional examinations for diagnosis, such as colonoscopy, and the evaluation of therapeutic responses, providing evidence of relapse or mucosal healing, which can lead to the intensification or reduction of treatment. As calprotectin levels are a measure of mucosal inflammation, high fecal concentrations are also found in other diseases with an inflammatory component, such as infectious enteritis or colorectal cancer. Interpretation of the concentration must therefore always take into account the clinical history and symptoms specific to each patient.
Collapse
Affiliation(s)
- Nathalie Kapel
- Laboratoire de Coprologie Fonctionnelle, Laboratoire de Biologie Médicale de Référence «Exploration Biochimique des Selles (Phénotype)», AP–HP, Hôpital Universitaire Pitié Salpêtrière-Charles Foix, Paris, France
- INSERM UMR-S1139, Faculté de Pharmacie, Université de Paris Cité, Paris, France
| | - Hamza Ouni
- Laboratoire de Coprologie Fonctionnelle, Laboratoire de Biologie Médicale de Référence «Exploration Biochimique des Selles (Phénotype)», AP–HP, Hôpital Universitaire Pitié Salpêtrière-Charles Foix, Paris, France
| | - Nacer Adam Benahmed
- Laboratoire de Coprologie Fonctionnelle, Laboratoire de Biologie Médicale de Référence «Exploration Biochimique des Selles (Phénotype)», AP–HP, Hôpital Universitaire Pitié Salpêtrière-Charles Foix, Paris, France
| | - Laurence Barbot-Trystram
- Laboratoire de Coprologie Fonctionnelle, Laboratoire de Biologie Médicale de Référence «Exploration Biochimique des Selles (Phénotype)», AP–HP, Hôpital Universitaire Pitié Salpêtrière-Charles Foix, Paris, France
| |
Collapse
|
10
|
Paysal J, Oris C, Troin U, Limeri PN, Allard J, Tadrent M, Pereira B, Merlin E, Rochette E, Evrard B, Durif J, Sapin V, Pons M. Influence of Perinatal Factors on Blood Tryptase and Fecal Calprotectin Levels in Newborns. CHILDREN (BASEL, SWITZERLAND) 2023; 10:345. [PMID: 36832475 PMCID: PMC9954950 DOI: 10.3390/children10020345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND Blood tryptase and fecal calprotectin levels may serve as biomarkers of necrotizing enterocolitis. However, their interpretation may be hindered by the little-known effects of perinatal factors. The aim of this study was to compare the tryptase and calprotectin levels in newborns according to their term, trophicity, and sex. METHOD One hundred and fifty-seven premature newborns and 157 full-term newborns were included. Blood tryptase and fecal calprotectin were assayed. RESULTS Blood tryptase levels were higher in premature than in full-term newborns (6.4 vs. 5.2 µg/L; p < 0.001). In situations of antenatal use of corticosteroids (p = 0.007) and non-exclusive use of human milk (p = 0.02), these levels were also higher. However, in multiple linear regression analyses, only prematurity significantly influenced tryptase levels. Fecal calprotectin levels were extremely wide-ranging and were much higher in female than in male newborns (300.5 vs. 110.5 µg/g; p < 0.001). CONCLUSIONS The differences in tryptase levels according to term could be linked to early aggression of the still-immature digestive wall in premature newborns, in particular, by enteral feeding started early. The unexpected influence of sex on fecal calprotectin levels remains unexplained.
Collapse
Affiliation(s)
- Justine Paysal
- CHU Clermont-Ferrand, Néonatologie et Réanimation Pédiatrique, F-63000 Clermont-Ferrand, France
- INSERM, CIC 1405, CRECHE Unit, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| | - Charlotte Oris
- CHU Clermont-Ferrand Gabriel Montpied, Biochimie, F-63000 Clermont-Ferrand, France
| | - Ugo Troin
- INSERM, CIC 1405, CRECHE Unit, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| | - Pierre-Nicolas Limeri
- INSERM, CIC 1405, CRECHE Unit, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| | - Jeanne Allard
- INSERM, CIC 1405, CRECHE Unit, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| | - Marie Tadrent
- INSERM, CIC 1405, CRECHE Unit, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| | - Bruno Pereira
- CHU Clermont-Ferrand, Direction de la Recherche Clinique et de l’Innovation, Secteur Biométrie et Médico-économi, F-63000 Clermont-Ferrand, France
| | - Etienne Merlin
- INSERM, CIC 1405, CRECHE Unit, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
- CHU Clermont-Ferrand, Pédiatrie, F-63000 Clermont-Ferrand, France
| | - Emmanuelle Rochette
- INSERM, CIC 1405, CRECHE Unit, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
- CHU Clermont-Ferrand, Pédiatrie, F-63000 Clermont-Ferrand, France
| | - Bertrand Evrard
- CHU Clermont-Ferrand, Immunologie, F-63000 Clermont-Ferrand, France
| | - Julie Durif
- CHU Clermont-Ferrand Gabriel Montpied, Biochimie, F-63000 Clermont-Ferrand, France
| | - Vincent Sapin
- CHU Clermont-Ferrand Gabriel Montpied, Biochimie, F-63000 Clermont-Ferrand, France
| | - Maguelonne Pons
- CHU Clermont-Ferrand, Chirurgie Pédiatrique, F-63000 Clermont-Ferrand, France
| |
Collapse
|
11
|
Lee WS, Arai K, Alex G, Treepongkaruna S, Kim KM, Choong CL, Mercado KC, Darma A, Srivastava A, Aw MM. Management and monitoring of pediatric inflammatory bowel disease in the Asia-Pacific region: A position paper by the Asian Pan-Pacific Society for Pediatric Gastroenterology, Hepatology, and Nutrition (APPSPGHAN) PIBD Working Group: Surgical management, disease monitoring, and special considerations. J Gastroenterol Hepatol 2022; 38:510-522. [PMID: 36508314 DOI: 10.1111/jgh.16084] [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] [Received: 09/01/2022] [Revised: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 01/15/2023]
Abstract
Disease phenotype of pediatric inflammatory bowel disease (PIBD) in children from the Asia-Pacific region differs from that of children from the West. Many parts of Asia are endemic for tuberculosis, making diagnosis and management of pediatric Crohn's disease a challenge. Current available guidelines, mainly from Europe and North America, may not be completely applicable to clinicians caring for children with PIBD in Asia due to differences in disease characteristics and regional resource constraints. This position paper is an initiative from the Asian Pan-Pacific Society for Pediatric Gastroenterology, Hepatology and Nutrition (APPSPGHAN) that aims to provide an up-to-date, evidence-based approach to PIBD in the Asia-Pacific region. A group of pediatric gastroenterologists with a special interest in PIBD performed an extensive literature search covering epidemiology, disease characteristics and natural history, management, and monitoring. Attention was paid to publications from the region with special consideration to a resource-limited setting. This current position paper deals with surgical management, disease monitoring, immunization, bone health, and nutritional issues of PIBD in Asia. A special section on differentiating pediatric Crohn's disease from tuberculosis in children is included. This position paper provides a useful guide to clinicians in the surgical management, disease monitoring, and various health issues in children with IBD in Asia-Pacific region.
Collapse
Affiliation(s)
- Way Seah Lee
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia.,Department of Population Medicine, Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, 59100, Kajang, Selangor, Malaysia
| | - Katsuhiro Arai
- Center for Pediatric Inflammatory Bowel Disease, National Center for Child Health and Development, Tokyo, Japan
| | - George Alex
- Department of Gastroenterology and Nutrition, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Suporn Treepongkaruna
- Division of Gastroenterology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kyung Mo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chee Liang Choong
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Karen Calixto Mercado
- Makati Medical Center and The Medical City, Philippine Society for Pediatric Gastroenterology, Hepatology and Nutrition, Manila, Philippines
| | - Andy Darma
- Department of Child Health, Dr. Soetomo General Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Anshu Srivastava
- Department of Paediatric Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Marion M Aw
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | |
Collapse
|
12
|
Trends of fecal calprotectin levels and associations with early life experience in preterm infants. INTERDISCIPLINARY NURSING RESEARCH 2022; 1:36-42. [PMID: 36590866 PMCID: PMC9766919 DOI: 10.1097/nr9.0000000000000006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/23/2022] [Indexed: 12/03/2022]
Abstract
Background Preterm infants are at risk for severe infections due to their immature immune systems. Factors such as early life pain/stress experiences and feeding may influence immune activation and maturation of immune systems. However, the underlying mechanism remains unclear. Fecal calprotectin (FCP) is a noninvasive surrogate biomarker of mucosal inflammation in the gastrointestinal tract and has been used in detecting intestinal inflammation in specific pediatric gastrointestinal disorders. Objective To describe the longitudinal trajectory of FCP levels in preterm infants and investigate the contributing factors that are associated with FCP levels. Design A longitudinal study design was used. Settings Preterm infants were recruited from 2 neonatal intensive care units (NICU) of a children's medical center in the North-eastern US. Methods Preterm infants were followed during their first 4 weeks of NICU hospitalization. Stool samples were collected twice per week to quantify the FCP levels. Cumulative pain/stress experiences and feeding types were measured daily. A linear mixed-effect model was used to examine the associations between FCP levels and demographic and clinical characteristics, cumulative pain/stress, and feeding over time. Results Forty-nine preterm infants were included in the study. Infants' FCP levels varied largely with a mean of 268.7±261.3 µg/g and increased over time. Preterm infants experienced an average of 7.5±5.0 acute painful procedures and 15.3±20.8 hours of chronic painful procedures per day during their NICU stay. The mean percentage of mother's own milk increased from the first week (57.1±36.5%) to the fourth week (60.7±38.9%) after birth. Elevated FCP concentration was associated with acute and cumulative (chronic) pain/stress levels, mother's own milk, non-White race, and higher severity of illness score. Conclusions FCP levels were elevated in preterm infants with wide interindividual and intraindividual variations. Cumulative pain/stress during the NICU hospitalization, feeding, race, and health status may influence FCP concentrations in early life that may be associated with inflammatory gut processes.
Collapse
|
13
|
Cekovic JR, Prodanovic NS, Mijailovic SS, Knezevic SM, Vuletic BP, Stojkovic AK, Savic DM, Prodanovic TV, Stanojevic MM, Simovic AM. The perinatal factors that influence the excretion of fecal calprotectin in premature-born children. Open Med (Wars) 2022; 17:1275-1281. [PMID: 35892079 PMCID: PMC9281588 DOI: 10.1515/med-2022-0522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/04/2022] [Accepted: 06/12/2022] [Indexed: 11/17/2022] Open
Abstract
This study aimed to provide additional information on the influence of perinatal factors on fecal (f)-calprotectin values in preterm infants. Calprotectin was determined from the first spontaneous stool (analyzed on the Alegria device by using the enzyme-linked immunosorbent assay [ELISA] method) obtained from neonates at a mean age of 3.41 ± 2.44 days of life. We analyzed 114 subjects who had a body weight of 1847.67 ± 418.6 g and were born at a gestational age of 32.6 ± 2.43 weeks, without intestinal and other congenital anomalies or any diseases other than those related to premature birth. The values of f-calprotectin are in a positive correlation with female subjects, intrauterine growth restriction, significant ductus arteriosus, enteral feeding intolerance, postnatal prolonged use of broad-spectrum antibiotics, and values of bicarbonates (analyzed in a sample of capillary arterial blood). Measurement of f-calprotectin in the first 7 days after birth can help to early detect the intestinal distress or early staging of necrotizing enterocolitis in premature infants.
Collapse
Affiliation(s)
- Jelena R. Cekovic
- Neonatal Intensive Care Unit, Center for Neonatology, Pediatric Clinic, University Clinical Centre Kragujevac, Kragujevac, Serbia
| | - Nikola S. Prodanovic
- Department of Alloartoplastic Surgery, Clinic for Orthopedics and Traumatology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Sara S. Mijailovic
- Department of Medical Statistics and Informatics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Sanja M. Knezevic
- Department of Pediatrics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Cardiology, Pediatric Clinic, University Clinical Centre Kragujevac, Kragujevac, Serbia
| | - Biljana P. Vuletic
- Department of Pediatrics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Gastroenterology, Pediatric Clinic, University Clinical Centre Kragujevac, Kragujevac, Serbia
| | - Andjelka K. Stojkovic
- Department of Pediatrics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Pulmonology, Pediatric Clinic, University Clinical Centre Kragujevac, Kragujevac, Serbia
| | - Dragana M. Savic
- Neonatal Intensive Care Unit, Center for Neonatology, Pediatric Clinic, University Clinical Centre Kragujevac, Kragujevac, Serbia
- Department of Pediatrics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tijana V. Prodanovic
- Neonatal Intensive Care Unit, Center for Neonatology, Pediatric Clinic, University Clinical Centre Kragujevac, Kragujevac, Serbia
| | - Marina M. Stanojevic
- Department of Neonatology, Clinic for Gynecology and Obstetrics, University Clinical Centre Kragujevac, Kragujevac, Serbia
| | - Aleksandra M. Simovic
- Neonatal Intensive Care Unit, Center for Neonatology, Pediatric Clinic, University Clinical Centre Kragujevac, Kragujevac, Serbia
- Department of Pediatrics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
14
|
Intestinal Inflammation is Significantly Associated With Length Faltering in Preterm Infants at Neonatal Intensive Care Unit Discharge. J Pediatr Gastroenterol Nutr 2022; 74:837-844. [PMID: 35442225 PMCID: PMC9296612 DOI: 10.1097/mpg.0000000000003455] [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: 12/10/2022]
Abstract
OBJECTIVE The aim of the study was to assess intestinal inflammatory measures, urinary intestinal fatty acid-binding protein (IFABP), and fecal calprotectin (FC) by gestational age (GA) and postmenstrual age (PMA) and determine the association between intestinal inflammation and growth in preterm infants from birth to hospital discharge. We hypothesized that intestinal inflammation is associated with adverse growth in preterm infants. METHODS We assayed repeated measures of IFABP and FC in 72 hospitalized preterm infants (<34 weeks' gestation). We calculated weight and length z scores at birth and discharge using the Fenton growth reference. Associations between mean IFABP or FC, growth z scores at discharge, and growth faltering (weight or length z score difference <-0.8 from birth to discharge) were assessed using mixed linear and logistic regression models, adjusted for intrafamilial correlation and potential confounders: GA, sex, birth z score, race/ethnicity, and maternal age. RESULTS Mean IFABP was greater among infants born at earlier GA and decreased with increasing PMA. Mean FC did not vary by GA or PMA. Higher mean IFABP and FC were associated with lower discharge growth z scores and greater likelihood of growth faltering significant only for mean IFABP and discharge length z score (β = -0.353, 95% confidence interval [CI]: -0.704 to -0.002) and mean IFABP and length faltering (odds ratio [OR] 1.99, P = 0.018). CONCLUSIONS Intestinal inflammation, measured by IFABP, was associated with lower length z scores and length faltering at discharge. Interventions to prevent intestinal inflammation may improve linear growth among preterm infants.
Collapse
|
15
|
Chen K, Zhang G, Xie H, You L, Li H, Zhang Y, Du C, Xu S, Melsaether C, Yuan S. Efficacy of Bifidobacterium animalis subsp. lactis, BB-12 ® on infant colic - a randomised, double-blinded, placebo-controlled study. Benef Microbes 2021; 12:531-540. [PMID: 34550055 DOI: 10.3920/bm2020.0233] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To evaluate the administration of Bifidobacterium animalis subsp. lactis, BB-12® (BB-12) on infant colic in breastfed infants, a double-blind, placebo-controlled randomised study was conducted in Chengdu, China from April 2016 to October 2017 with 192 full-term infants less than 3 months of age and meeting the ROME III criteria for infant colic. After a 1-week run-in the infants were randomly assigned to receive daily BB-12 (1×109 cfu/day) or placebo for 3 weeks. Crying/fussing time were recorded using a 24 h structured diary. The primary endpoint was the proportion of infants achieving a reduction in crying and fussing time of ≥50% from baseline. Parent's/caregiver's health related quality of life was measured using a modified PedsQL™ 2.0 Family Impact Module and immunological biomarkers were evaluated from faecal samples at baseline and after the 21-day intervention. The percentage of infants achieving a reduction in the daily crying/fussing time ≥50% after the 21-day intervention was significantly higher in the infants supplemented with BB-12 (P<0.001). The mean number of crying episodes was significantly reduced in the BB-12 group compared to the placebo group (10.0±3.0 to 5.0±1.87 vs 10.5±2.6 to 7.5±2.8, respectively) (P<0.001) and the mean daily sleep duration was markedly increased from baseline to end of intervention in the BB-12 group compared to the infants in the placebo group (60.7±104.0 vs 31.9±102.7 min/day, respectively) (P<0.001). The faecal levels of human beta defensin 2, cathelicidin, slgA, calprotectin and butyrate were statistically higher in the BB-12 group compared to the placebo group after the 21-day intervention. At the end of the intervention the parent's/caregiver's physical, emotional and social functioning scores were significantly higher for the BB-12 group compared to the placebo group (all P<0.05). Supplementation of BB-12 is effective in reducing crying and fussing in infants diagnosed with infant colic.
Collapse
Affiliation(s)
- K Chen
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, 6100131, China P.R.,Department of Child Health Care, Chengdu New Century Women's and Children's Hospital, No.77, Baojia Lane, Qingyang District, Chengdu, China P.R
| | - G Zhang
- Department of Pediatric Intensive Care Unit, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, China P.R
| | - H Xie
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, No. 539, Inner Mongolia Avenue, Jinyuan Town, Dayi County, Chengdu, China P.R
| | - L You
- Department of Child Health Care, Nanxin Community Health Service Center, N0. 168, Guanghe 1st Street, Wuhou District, Chengdu, China P.R
| | - H Li
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Care Hospital, No.87, Qingjiang South Road, Qingbaijiang District, Chengdu, China P.R
| | - Y Zhang
- Department of Child Health Care, Jinniu Maternal and Child Health Care Hospital, No.12, Changyue Road, Jinniu District, Chengdu, China P.R
| | - C Du
- Department of Child Health Care, Longquanyi Maternal and Child Health Care Hospital, No.383, Yuyang Road, Longquanyi District, Chengdu, China P.R
| | - S Xu
- Department of Child Health Care, Huili Maternal and Child Health Care Hospital, No. 41, Jindai Road West Section, Guoyuan Township, Huili County, Xichang, China P.R
| | - C Melsaether
- Chr. Hansen A/S, HH Clinical Development, Kogle Alle 6, 2970 Hoersholm, Denmark
| | - S Yuan
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, 6100131, China P.R
| |
Collapse
|
16
|
D’Auria E, Salvatore S, Acunzo M, Peroni D, Pendezza E, Di Profio E, Fiore G, Zuccotti GV, Verduci E. Hydrolysed Formulas in the Management of Cow's Milk Allergy: New Insights, Pitfalls and Tips. Nutrients 2021; 13:2762. [PMID: 34444922 PMCID: PMC8401609 DOI: 10.3390/nu13082762] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022] Open
Abstract
An allergy to cow's milk requires the avoidance of cow's milk proteins and, in some infants, the use of a hypoallergenic formula. This review aims to summarize the current evidence concerning different types of hydrolysed formulas (HF), and recommendations for the treatment of IgE- and non-IgE-mediated cow's milk allergy and functional gastrointestinal disorders in infancy, for which some dietary intervention and HF may be of benefit to both immune and motor mechanisms. Current guidelines recommend cow's milk protein (i.e., whey or casein) extensively hydrolysed formula (eHF) as the first choice for cow's milk allergy treatment, and amino acid formulas for more severe cases or those with reactions to eHF. Rice hydrolysed formulas (rHF) have also become available in recent years. Both eHF and rHF are well tolerated by the majority of children allergic to cow's milk, with no concerns regarding body growth or adverse effects. Some hydrolysates may have a pro-active effect in modulating the immune system due to the presence of small peptides and additional components, like biotics. Despite encouraging results on tolerance acquisition, evidence is still not conclusive, thus hampering our ability to draw firm conclusions. In clinical practice, the choice of hypoallergenic formula should be based on the infant's age, the severity, frequency and persistence of symptoms, immune phenotype, growth pattern, formula cost, and in vivo proof of tolerance and efficacy.
Collapse
Affiliation(s)
- Enza D’Auria
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Silvia Salvatore
- Department of Pediatrics, Ospedale “F. Del Ponte”, University of Insubria, 21100 Varese, Italy;
| | - Miriam Acunzo
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Diego Peroni
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, 56126 Pisa, Italy;
| | - Erica Pendezza
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (M.A.); (E.P.); (E.D.P.); (G.F.); (G.V.Z.); (E.V.)
| |
Collapse
|
17
|
The Use of Fecal Calprotectin Testing in Paediatric Disorders: A Position Paper of the European Society for Paediatric Gastroenterology and Nutrition Gastroenterology Committee. J Pediatr Gastroenterol Nutr 2021; 72:617-640. [PMID: 33716293 DOI: 10.1097/mpg.0000000000003046] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The aim of the study was to review the evidence regarding the clinical use and value of fecal calprotectin (FC) measurements in different gastrointestinal disorders in children. METHODS A literature search was conducted in the PubMed, MEDLINE, EMBASE, and Cochrane databases until October 31, 2019. Subtopics were identified and each assigned to individual authors. RESULTS A total of 28 recommendations were voted on using the nominal voting technique. Recommendations are given related to sampling, measurement methods, and results interpretation. The 14 authors anonymously voted on each recommendation using a 9-point scale (1 strongly disagree to 9 fully agree). Consensus was considered achieved if at least 75% of the authors voted 6, 7, 8, or 9. CONCLUSIONS Consensus was reached for all recommendations. Limitations for the use of FC in clinical practice include variability in extraction methodology, performance of test kits as well as the need to establish local reference ranges because of the influence of individual factors, such as age, diet, microbiota, and drugs. The main utility of FC measurement at present is in the diagnosis and monitoring of inflammatory bowel disease (IBD) as well as to differentiate it from functional gastrointestinal disorders (FAPDs). FC, however, has neither utility in the diagnosis of infantile colic nor to differentiate between functional and organic constipation. A rise in FC concentration, may alert to the risk of developing necrotizing enterocolitis and help identifying gastrointestinal involvement in children with Henoch-Schönlein purpura. FC measurement is of little value in Cow's Milk Protein Allergy, coeliac disease (CD), and cystic fibrosis. FC does neither help to distinguish bacterial from viral acute gastroenteritis (AGE), nor to diagnose Helicobacter Pylori infection, small intestinal bacterial overgrowth (SIBO), acute appendicitis (AA), or intestinal polyps.
Collapse
|
18
|
Kim ES, Tarassishin L, Eisele C, Barre A, Nair N, Rendon A, Hawkins K, Debebe A, White S, Thjømøe A, Mørk E, Bento-Miranda M, Panchal H, Agrawal M, Patel A, Chen CL, Kornbluth A, George J, Legnani P, Maser E, Loudon H, Mella MT, Stone J, Dubinsky M, Sabino J, Torres J, Colombel JF, Peter I, Hu J. Longitudinal Changes in Fecal Calprotectin Levels Among Pregnant Women With and Without Inflammatory Bowel Disease and Their Babies. Gastroenterology 2021; 160:1118-1130.e3. [PMID: 33307026 DOI: 10.1053/j.gastro.2020.11.050] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/09/2020] [Accepted: 11/29/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The effect of pregnancy on inflammatory bowel disease (IBD) remains poorly understood. We aimed to monitor intestinal inflammation using fecal calprotectin (FC) in pregnant women and their babies during early life. METHODS Pregnant women with or without IBD and their infants were prospectively enrolled. FC levels were measured at each trimester of pregnancy and in babies throughout the first 3 years of life. Repeated-measures analysis was applied to investigate changes in FC levels while adjusting for confounders. The FC levels were correlated with the bacterial abundance in both mothers and babies. RESULTS Six hundred and fourteen fecal samples from 358 mothers (98 with IBD) and 1005 fecal samples from 289 infants (76 born to IBD mothers) were analyzed. Pregnant Patients with IBD maintained higher FC levels through pregnancy compared with controls (P = 7.5 × 10-54). FC gradually increased in controls and declined in Patients with IBD throughout pregnancy (P for interaction = 5.8 × 10-7). Babies born to mothers with IBD presented with significantly higher FC levels than those born to controls up to 3 years of age, after adjusting for sex, delivery mode, feeding behavior, and antibiotics exposure (2 weeks to 3 months of age, P = .015; 12-36 months of age, P = .00003). Subdoligranulum, Roseburia, Fusicatenibacter, and Alistipes negatively correlated, and Streptococcus, Prevotella, Escherichia-Shigella, and Bifidobacterium positively correlated with maternal FC levels at T3. Faecalibacterium, Bifidobacterium, and Alistipes showed negative correlations, and Streptococcus were positively correlated with FC levels within 3 months of birth. CONCLUSIONS Pregnancy is associated with decreased inflammatory activity in mothers with IBD. Higher FC levels in babies born to mothers with IBD suggest subclinical inflammation in early life, the long-term consequences of which are uncertain.
Collapse
Affiliation(s)
- Eun Soo Kim
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Gastroenterology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Leonid Tarassishin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Caroline Eisele
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amelie Barre
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Gastroenterology, Cochin Hospital, Université de Paris, Paris, France
| | - Nilendra Nair
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Alexa Rendon
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kelly Hawkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anketse Debebe
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sierra White
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Mario Bento-Miranda
- Division of Gastroenterology, Hospital and University Center of Coimbra, Coimbra, Portugal
| | - Hinaben Panchal
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Manasi Agrawal
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anish Patel
- Division of Gastroenterology, Brooke Army Medical Center, San Antonio, Texas
| | - Ching-Lynn Chen
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Asher Kornbluth
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James George
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter Legnani
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elana Maser
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Holly Loudon
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Maria-Teresa Mella
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joanne Stone
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marla Dubinsky
- Department of Pediatric Gastroenterology and Nutrition, Icahn School of Medicine at Mount Sinai, New York, New York
| | - João Sabino
- Department of Gastroenterology, University Hospitals of Leuven, Leuven, Belgium
| | - Joana Torres
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Gastroenterology, Surgical Department, Hospital Beatriz Ângelo, Loures, Portugal
| | - Jean-Frederic Colombel
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Inga Peter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jianzhong Hu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| | | |
Collapse
|
19
|
Guidance on the interpretation of faecal calprotectin levels in children. PLoS One 2021; 16:e0246091. [PMID: 33571226 PMCID: PMC7877663 DOI: 10.1371/journal.pone.0246091] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/13/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Faecal calprotectin (FCP) is a powerful tool to predict inflammatory bowel disease (IBD) in patients with gastrointestinal symptoms. In the paediatric patient population, the reference value of < 50 μg/g and the influence of age on FCP levels result in a high number of redundant investigations and specialist referrals. We assessed paediatric FCP levels, their diagnostic value and corresponding referral pathways from primary and secondary care. METHODS We analysed two cohorts from a precisely defined catchment area: one consisted of all FCPs measured in this area (n = 2788). The second cohort-a subset of the first cohort-consisted of FCP values and corresponding clinical data from children who were referred for possible IBD to our department (n = 373). RESULTS In the first cohort, 47% of FCP levels were > 50 μg/g, 15% were ≥ 250 μg/g. Children < 1y had significantly (p < 0.001) higher FCP than older children. In the second cohort, 6.7% of children with an FCP of < 250 μg/g (or 8.6% with an FCP of < 600 μg/g) had IBD-all featured symptoms suggestive of IBD (e.g. bloody diarrhoea, nocturnal abdominal pain, weight loss) or abnormal blood tests. 76% of patients in whom raised FCP (> 50 μg/g) was the sole reason for being referred for suspected IBD did not have IBD. CONCLUSION Children with an FCP < 600 μg/g and without matching symptoms suggestive of IBD are unlikely to have IBD. A higher FCP reference value may provide cost-effective improvement that could avoid redundant investigations and specialist referrals. A guideline for specialist referrals is proposed.
Collapse
|
20
|
Campeotto F, Elie C, Rousseau C, Giuseppi A, Hachem T, Gobalakichenane P, Le Touzey M, Stefano M, Butel M, Kapel N. Faecal calprotectin and gut microbiota do not predict enteropathy in very preterm infants. Acta Paediatr 2021; 110:109-116. [PMID: 32418251 PMCID: PMC7818215 DOI: 10.1111/apa.15354] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
Aim Very preterm birth is associated with a high risk of enteropathies. Diagnosis is challenging, especially in mild forms, leading to unnecessary periods of cessation of enteral feeding. This study aimed at establishing a prognosis score of enteropathy combining clinical parameters and faecal calprotectin concentration. Methods This prospective multicentric study included preterm neonates born at a gestational age of 33 weeks or less. Stools were collected weekly until hospital discharge, and daily in case of digestive events for calprotectin measurement (ELISA and immunochromatography) and microbiota analyses (16S rRNA gene sequencing). Results Among the 121 neonates included, 21 experienced at least one episode of enteropathy, mainly mild forms. By ELISA testing, median faecal calprotectin was 88 (8‐798) µg/g faeces. No statistically significant association was found between the outset of enteropathy and maternal and neonatal characteristics, and calprotectin levels. The agreement between ELISA and immunochromatography assay was moderate (intra‐class correlation coefficient 0.58, 95%CI [0.47‐0.66]). Comparison of species diversity and relative bacterial abundance profiles between infants with or without enteropathy revealed no specific alterations associated with enteropathy. Conclusion The study failed to propose a prognostic score of enteropathy, probably due the large inter‐ and intra‐individual variability of faecal calprotectin in very preterm neonates.
Collapse
Affiliation(s)
- Florence Campeotto
- Department of Pediatric Gastroenterology Necker‐Enfants Malades HospitalAP‐HP Paris France
- Faculty of Pharmacy INSERMU1139 Paris France
- UMR‐S U1139 Hospital‐University Department Risks In PregnancyParis Descartes UniversityParis University Paris France
| | - Caroline Elie
- Clinical Research Unit Clinical Investigation Center Necker‐Enfants Malades HospitalAP‐HP Paris France
| | - Clotilde Rousseau
- Faculty of Pharmacy INSERMU1139 Paris France
- UMR‐S U1139 Hospital‐University Department Risks In PregnancyParis Descartes UniversityParis University Paris France
- Microbiology Department St‐Louis HospitalAPHP Paris France
| | - Agnès Giuseppi
- Neonatology Department Necker‐Enfants Malades HospitalAP‐HP Paris France
| | - Taymme Hachem
- Neonatology Department Necker‐Enfants Malades HospitalAP‐HP Paris France
| | | | - Mathilde Le Touzey
- Neonatology Department Poissy‐Saint Germain Intercommunal Hospital Center Poissy France
| | - Marie Stefano
- Faculty of Pharmacy INSERMU1139 Paris France
- UMR‐S U1139 Hospital‐University Department Risks In PregnancyParis Descartes UniversityParis University Paris France
| | - Marie‐José Butel
- Faculty of Pharmacy INSERMU1139 Paris France
- UMR‐S U1139 Hospital‐University Department Risks In PregnancyParis Descartes UniversityParis University Paris France
| | - Nathalie Kapel
- Faculty of Pharmacy INSERMU1139 Paris France
- UMR‐S U1139 Hospital‐University Department Risks In PregnancyParis Descartes UniversityParis University Paris France
- Coprology Department Pitié‐Salpétrière HospitalAPHP Paris France
| |
Collapse
|
21
|
Günaydın Şahin BS, Keskindemirci G, Özden TA, Durmaz Ö, Gökçay G. Faecal calprotectin levels during the first year of life in healthy children. J Paediatr Child Health 2020; 56:1806-1811. [PMID: 32502317 DOI: 10.1111/jpc.14933] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/01/2022]
Abstract
AIM A high faecal calprotectin (FC) level is a non-invasive marker for inflammatory bowel disease. Nevertheless, healthy infants have elevated levels of FC with large variations. The aim of our study was to determine the levels of FC and associated factors in healthy infants aged 0-12 months. METHODS Infants younger than 1 year of age were in the follow-up programme of the Well Child Unit. Data on the clinical characteristics, including birth, anthropometric measurements and feeding types of infants in the unit, were obtained from their personal health records. One fresh stool sample was collected from each infant. ELISA was used to measure FC. RESULTS We included 84 infants younger than 1 year of age. The median FC value was 313 μg/g. The FC levels were greater in the youngest (0-30 days) group of infants than in the oldest (181-365 days) group (P < 0.001). The FC levels were higher in infants delivered by caesarean section than in those delivered vaginally (P = 0.016). The levels were also higher in infants who were solely breastfed than in those who received mixed feeding (breast milk and formula) during the first 6 months of life (P = 0.030). CONCLUSION The FC levels in this group of infants were high, especially in the first month of life. Several birth and environmental factors influenced the FC values. Further studies with a larger cohort of infants and serial assessment of FC over time are required to better understand the patterns of this biomarker during infancy.
Collapse
Affiliation(s)
| | - Gonca Keskindemirci
- Division of Social Paediatrics, Department of Paediatrics, İstanbul Medical Faculty, İstanbul University, İstanbul, Turkey.,Institute of Health Sciences, and Institute of Child Health, Social Paediatrics PhD Program, İstanbul University, İstanbul, Turkey
| | - Tülin Ayşe Özden
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, İstanbul Medical Faculty, İstanbul University, İstanbul, Turkey
| | - Özlem Durmaz
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, İstanbul Medical Faculty, İstanbul University, İstanbul, Turkey
| | - Gülbin Gökçay
- Division of Social Paediatrics, Department of Paediatrics, İstanbul Medical Faculty, İstanbul University, İstanbul, Turkey.,Department of Social Paediatrics, Institute of Child Health, İstanbul University, Fatih, İstanbul, Turkey
| |
Collapse
|
22
|
Shores DR, Fundora J, Go M, Shakeel F, Brooks S, Alaish SM, Yang J, Sodhi CP, Hackam DJ, Everett A. Normative values for circulating intestinal fatty acid binding protein and calprotectin across gestational ages. BMC Pediatr 2020; 20:250. [PMID: 32456678 PMCID: PMC7249444 DOI: 10.1186/s12887-020-02142-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/12/2020] [Indexed: 01/31/2023] Open
Abstract
Background Necrotizing enterocolitis (NEC) is associated with significant morbidity and mortality. Serum biomarkers to aid diagnosis, such as intestinal fatty acid binding protein (IFABP) and calprotectin, are actively being investigated; however, the normative values of these markers among healthy premature and term infants remains unknown. We sought to identify normative values for the serum concentrations of IFABP and calprotectin across gestational (GA) and post-menstrual age. Methods We collected serum from infants (24–40 weeks GA) in the first week of life and at multiple time points in a sub-cohort of premature infants (24–29 weeks GA), excluding sepsis or known intestinal disease. IFABP and calprotectin were measured using ELISA. Groups were compared with descriptive statistics and mixed effects linear regression. Results One hundred twelve infants had specimens in the first week of life, and 19 premature infants had longitudinal specimens. IFABP concentration in the first week of life was low and did not differ across gestational ages. Longitudinally, IFABP increased 4% per day (P < 0.001). Calprotectin concentration in the first week of life was more variable. An inverse relationship between day of life and calprotectin level was found in the longitudinal cohort (P < 0.001). Conclusions Serum IFABP and calprotectin fluctuate over time. Infants had low levels of IFABP during the first week of life, independent of gestational age, and levels increased longitudinally in premature infants. Calprotectin levels generally declined over time. Normative data for infants is necessary to establish meaningful cut-off levels for clinical use.
Collapse
Affiliation(s)
- Darla R Shores
- Department of Pediatrics, Division of Gastroenterology, Hepatology, & Nutrition, Johns Hopkins School of Medicine, 600 N. Wolfe St, CMSC 2-116, Baltimore, MD, 21187, USA.
| | - Jennifer Fundora
- Department of Pediatrics, Johns Hopkins Hospital, 1800 Orleans St, Ste 8520, Baltimore, MD, 21287, USA
| | - Mitzi Go
- Department of Pediatrics, Division of Neonatology, Johns Hopkins Medicine All Children's Hospital, 600 5th Street South, St. Petersburg, FL, 33701, USA
| | - Fauzia Shakeel
- Department of Pediatrics, Division of Maternal Fetal and Neonatal Institute Johns Hopkins All Children's Hospital, 601 5th Street South, St. Petersburg, FL, 33701, USA
| | - Sandra Brooks
- Department of Pediatrics, Division of Neonatology, Johns Hopkins Medicine All Children's Hospital, 600 5th Street South, St. Petersburg, FL, 33701, USA
| | - Samuel M Alaish
- Department of Surgery, Division of General Pediatric Surgery, Johns Hopkins School of Medicine, 1800 Orleans Street, Room 7337, Baltimore, MD, 21287, USA
| | - Jun Yang
- Department of Pediatrics, Division of Cardiology, Johns Hopkins School of Medicine, 720 Rutland Ave, Ross 1143, Baltimore, MD, 21205, USA
| | - Chhinder P Sodhi
- Department of Surgery, Division of General Pediatric Surgery, Johns Hopkins School of Medicine, 1800 Orleans Street, Room 7337, Baltimore, MD, 21287, USA
| | - David J Hackam
- Department of Surgery, Division of General Pediatric Surgery, Johns Hopkins School of Medicine, 1800 Orleans Street, Room 7337, Baltimore, MD, 21287, USA
| | - Allen Everett
- Department of Pediatrics, Division of Cardiology, Johns Hopkins School of Medicine, 720 Rutland Ave, Ross 1143, Baltimore, MD, 21205, USA
| |
Collapse
|
23
|
Early Effect of Supplemented Infant Formulae on Intestinal Biomarkers and Microbiota: A Randomized Clinical Trial. Nutrients 2020; 12:nu12051481. [PMID: 32443684 PMCID: PMC7284641 DOI: 10.3390/nu12051481] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/11/2020] [Accepted: 05/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Post-natal gut maturation in infants interrelates maturation of the morphology, digestive, and immunological functions and gut microbiota development. Here, we explored both microbiota development and markers of gut barrier and maturation in healthy term infants during their early life to assess the interconnection of gut functions during different infant formulae regimes. Methods: A total of 203 infants were enrolled in this randomized double-blind controlled trial including a breastfed reference group. Infants were fed starter formulae for the first four weeks of life, supplemented with different combination of nutrients (lactoferrin, probiotics (Bifidobacterium animal subsp. Lactis) and prebiotics (Bovine Milk-derived Oligosaccharides—BMOS)) and subsequently fed the control formula up to eight weeks of life. Stool microbiota profiles and biomarkers of early gut maturation, calprotectin (primary outcome), elastase, α-1 antitrypsin (AAT) and neopterin were measured in feces at one, two, four, and eight weeks. Results: Infants fed formula containing BMOS had lower mean calprotectin levels over the first two to four weeks compared to the other formula groups. Elastase and AAT levels were closer to levels observed in breastfed infants. No differences were observed for neopterin. Global differences between the bacterial communities of all groups were assessed by constrained multivariate analysis with hypothesis testing. The canonical correspondence analysis (CCA) at genus level showed overlap between microbiota profiles at one and four weeks of age in the BMOS supplemented formula group with the breastfed reference, dominated by bifidobacteria. Microbiota profiles of all groups at four weeks were significantly associated with the calprotectin levels at 4 (CCA, p = 0.018) and eight weeks of age (CCA, p = 0.026). Conclusion: A meaningful correlation was observed between changes in microbiota composition and gut maturation marker calprotectin. The supplementation with BMOS seems to favor gut maturation closer to that of breastfed infants.
Collapse
|
24
|
Beneficial Effect of Mildly Pasteurized Whey Protein on Intestinal Integrity and Innate Defense in Preterm and Near-Term Piglets. Nutrients 2020; 12:nu12041125. [PMID: 32316586 PMCID: PMC7230795 DOI: 10.3390/nu12041125] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 02/08/2023] Open
Abstract
Background. The human digestive tract is structurally mature at birth, yet maturation of gut functions such as digestion and mucosal barrier continues for the next 1–2 years. Human milk and infant milk formulas (IMF) seem to impact maturation of these gut functions differently, which is at least partially related to high temperature processing of IMF causing loss of bioactive proteins and formation of advanced glycation end products (AGEs). Both loss of protein bioactivity and formation of AGEs depend on heating temperature and time. The aim of this study was to investigate the impact of mildly pasteurized whey protein concentrate (MP-WPC) compared to extensively heated WPC (EH-WPC) on gut maturation in a piglet model hypersensitive to enteral nutrition. Methods. WPC was obtained by cold filtration and mildly pasteurized (73 °C, 30 s) or extensively heat treated (73 °C, 30 s + 80 °C, 6 min). Preterm (~90% gestation) and near-term piglets (~96% gestation) received enteral nutrition based on MP-WPC or EH-WPC for five days. Macroscopic and histologic lesions in the gastro-intestinal tract were evaluated and intestinal responses were further assessed by RT-qPCR, immunohistochemistry and enzyme activity analysis. Results. A diet based on MP-WPC limited epithelial intestinal damage and improved colonic integrity compared to EH-WPC. MP-WPC dampened colonic IL1-β, IL-8 and TNF-α expression and lowered T-cell influx in both preterm and near-term piglets. Anti-microbial defense as measured by neutrophil influx in the colon was only observed in near-term piglets, correlated with histological damage and was reduced by MP-WPC. Moreover, MP-WPC stimulated iALP activity in the colonic epithelium and increased differentiation into enteroendocrine cells compared to EH-WPC. Conclusions. Compared to extensively heated WPC, a formula based on mildly pasteurized WPC limits gut inflammation and stimulates gut maturation in preterm and near-term piglets and might therefore also be beneficial for preterm and (near) term infants.
Collapse
|
25
|
Łoniewska B, Adamek K, Węgrzyn D, Kaczmarczyk M, Skonieczna-Żydecka K, Clark J, Adler G, Tousty J, Uzar I, Tousty P, Łoniewski I. Analysis of Faecal Zonulin and Calprotectin Concentrations in Healthy Children During the First Two Years of Life. An Observational Prospective Cohort Study. J Clin Med 2020; 9:jcm9030777. [PMID: 32178435 PMCID: PMC7141325 DOI: 10.3390/jcm9030777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Factors affecting the intestinal-barrier permeability of newborns, such as body mass index (BMI), nutrition and antibiotics, are assumed to affect intestinal-barrier permeability in the first two years of life. This study assessed 100 healthy, full-term newborns to 24 months old. Faecal zonulin/calprotectin concentrations were measured at 1, 6, 12, 24 months as gut-permeability markers. Zonulin concentrations increased between 1 and 12 months (medians: 114.41, 223.7 ng/mL; respectively), whereas calprotectin concentrations decreased between one and six months (medians: 149. 29, 109.28 µg/mL); both then stabilized (24 months: 256.9 ng/mL zonulin; 59.5 µg/mL calprotectin). In individual children, high levels at one month gave high levels at older ages (correlations: calprotectin: between 1 and 6 or 12 months: correlation coefficient (R) = 0.33, statistical significance (p) = 0.0095; R = 0.28, p = 0.032; zonulin: between 1 and 24 months: R = 0.32; p = 0.022, respectively). Parameters which gave marker increases: antibiotics during pregnancy (calprotectin; six months: by 80%, p = 0.038; 12 months: by 48%, p = 0.028); vaginal birth (calprotectin: 6 months: by 140%, p = 0.005); and > 5.7 pregnancy-BMI increase (zonulin: 12 months: by 74%, p = 0.049). Conclusions: “Closure of the intestines” is spread over time and begins between the sixth and twelfth month of life. Antibiotic therapy, BMI increase > 5.7 during pregnancy and vaginal birth are associated with increased intestinal permeability during the first two years of life. Stool zonulin and calprotectin concentrations were much higher compared with previous measurements at older ages; clinical interpretation and validation are needed (no health associations found).
Collapse
Affiliation(s)
- Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
- Correspondence: ; Tel.: +48-(91)-466-1375
| | - Karolina Adamek
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Dagmara Węgrzyn
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin 70-111, Poland; (M.K.); (J.C.)
| | - Karolina Skonieczna-Żydecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin 71-460, Poland; (K.S.-Ż.); (I.Ł.)
| | - Jeremy Clark
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin 70-111, Poland; (M.K.); (J.C.)
| | - Grażyna Adler
- Department of Studies in Anthropogenetics and Biogerontology, Pomeranian Medical University, Szczecin 71-210, Poland;
| | - Joanna Tousty
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Izabela Uzar
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University, Szczecin 71-230, Poland;
| | - Piotr Tousty
- Department of Obstetrics and Gynecology, Pomeranian Medical University, Szczecin 70-111, Poland;
| | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin 71-460, Poland; (K.S.-Ż.); (I.Ł.)
| |
Collapse
|
26
|
Parker LA, Weaver M, Torrazza RJM, Shuster J, Li N, Krueger C, Neu J. Effect of Aspiration and Evaluation of Gastric Residuals on Intestinal Inflammation, Bleeding, and Gastrointestinal Peptide Level. J Pediatr 2020; 217:165-171.e2. [PMID: 31757473 PMCID: PMC7012706 DOI: 10.1016/j.jpeds.2019.10.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/01/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine the effect of gastric residual aspiration and evaluation on preterm very low birth weight infants' gastrointestinal function, intestinal inflammation, and gastrointestinal mucosal bleeding. STUDY DESIGN This single-center, randomized trial compared omission of gastric residuals vs prefeed gastric residuals in 143 infants ≤32 weeks of gestation with a birthweight of ≤1250 g for 6 weeks after birth. Serum levels of gastrin and motilin were collected between 14 and 21 days of life. Stools were collected at 3 and 6 weeks of age and analyzed for calprotectin and S100A12 levels. All stools were tested for occult blood for 6 weeks. RESULTS Means for gastrin (P = .999) and motilin (P = .694) were similar between groups and there were no statistically significant differences in adjusted means for transformed calprotectin (P = .580), and S100A12 (P = .212). Both calprotectin (P = .003) and S100A12 (P = .002) increased from week 3 to week 6. The mean percentage of stools positive for occult blood (P = .888) were similar between the groups. CONCLUSIONS Gastrointestinal function, intestinal inflammation, and gastrointestinal mucosal bleeding were similar whether aspiration and evaluation of gastric residuals were eliminated or not, suggesting routinely evaluating gastric residuals before every feeding may be unnecessary. TRIAL REGISTRATION CLINICALTRIALS.GOV:: NCT01863043.
Collapse
Affiliation(s)
- Leslie A. Parker
- College of Nursing, University of Florida; No conflict of interest
| | - Michael Weaver
- College of Nursing, University of Florida; No conflict of interest
| | - Roberto J. Murgas Torrazza
- Sistema Nacional de Investigacion de Panama (SNI). Secretaria Nacional de Ciencia Tecnologia e Innovacion (SENACYT); No conflict of interest
| | - Jonathon Shuster
- Department of Health Outcomes and Bioinformatics, University of Florida; No conflict of interest
| | - Nan Li
- Department of Pediatrics, University of Florida; No conflict of interest
| | - Charlene Krueger
- College of Nursing, University of Florida; No conflict of interest
| | - Josef Neu
- Department of Pediatrics, University of Florida; Conflict: Research grant from Infant Bacterial Therapeutics
| |
Collapse
|
27
|
Costa S, Patti ML, Perri A, Cocca C, Pinna G, Tirone C, Tana M, Lio A, Vento G. Effect of Different Milk Diet on the Level of Fecal Calprotectin in Very Preterm Infants. Front Pediatr 2020; 8:552. [PMID: 33042911 PMCID: PMC7524876 DOI: 10.3389/fped.2020.00552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/30/2020] [Indexed: 12/04/2022] Open
Abstract
Objective: To evaluate the course of fecal calprotectin (FC) in very preterm infants over the first 15 days of life in relation to the type of milk diet. Methods: This study was part of a randomized controlled trial comparing two different ways of integrating the own mother's milk (OMM) for the evaluation of feeding tolerance in very preterm infants. In infants with gestational age of ≤ 32 weeks randomized to receive preterm formula (PF group) or pasteurized donor human milk (PDHM group) as a supplement to the OMM insufficient or unavailable, FC level was planned to be measured at the first meconium passage and at days 8 and 15 of life (T0, T1, and T2, respectively). Results: FC data were available for all the 70 infants randomized, 35 in the PF group, and 35 in the PDHM group. The mean FC levels were similar in the two study groups at T0 and T1, whereas they were significantly higher in the PF group than the PDHM group at T2. FC values decreased over the first week of life in both groups and significantly increased over the second week of life only in the PF group. Conclusions: Our study demonstrates a significant increase in FC levels when PF is used as a supplement to the OMM compared to the use of PDHM. Further studies are needed to establish if the higher FC levels in infants receiving PF are the expression of a normal immunological maturation rather than an initial inflammatory process.
Collapse
Affiliation(s)
- Simonetta Costa
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Letizia Patti
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Perri
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carmen Cocca
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Pinna
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara Tirone
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Milena Tana
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandra Lio
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Vento
- Department of Woman and Child Health and Public Health, Child Health Area, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
28
|
Park JS, Cho JY, Chung C, Oh SH, Do HJ, Seo JH, Lim JY, Park CH, Woo HO, Youn HS. Dynamic Changes of Fecal Calprotectin and Related Clinical Factors in Neonates. Front Pediatr 2020; 8:326. [PMID: 32733824 PMCID: PMC7360719 DOI: 10.3389/fped.2020.00326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/19/2020] [Indexed: 11/13/2022] Open
Abstract
Objective: Fecal calprotectin (FC) has been widely used for a clinical marker of intestinal inflammation in children and adults. However, the clinical usefulness has not been determined in neonates. The purpose of this study was to investigate the change of FC and associated clinical factors in neonates. Methods and Materials: In total, 146 neonates among 472 admissions to our NICU between 2018 and 2019 were included, and 242 stool samples were collected. FC was measured in the first, second, and third-fourth week after birth, respectively, using commercial ELISA. The clinical characteristics were reviewed from medical records. Statistical analyses were performed to analyze associated factors regarding on changes of fecal calprotectin. Results: A wide range from 5.5 to 6,000 mg/kg of FC was observed in neonates. FCs during neonatal period were not correlated with the gestational age at birth or birth weight. The meconial calprotectin was higher than FCs after 2 weeks of age (n = 134, 418.06 vs. 243.12 in the second week and 259.58 in the third week after birth). Meconial calprotectin was associated with birth weight and meconium stained amniotic fluid. FC during the neonatal period decreased with postnatal week (-464.93 ± 158.02 at third-fourth week after birth compared with the 1st week, P = 0.004) and breast milk (-337.27 ± 150.51 compared with formula milk, P = 0.026). Conclusion: Fecal calprotectin tended to decrease with postnatal week during the neonatal period, and breast milk could affect more decrease of FC.
Collapse
Affiliation(s)
- Ji Sook Park
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Hospital, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Jae Young Cho
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Hospital, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Changyeong Chung
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Hospital, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Seong Hee Oh
- Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Hyun-Jeong Do
- Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Ji-Hyun Seo
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Hospital, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Jae Young Lim
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Hospital, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Chan-Hoo Park
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Hyang-Ok Woo
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Hospital, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Hee-Shang Youn
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, South Korea.,Department of Pediatrics, Gyeongsang National University Hospital, Jinju, South Korea.,Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
29
|
D'Auria E, Salvatore S, Pozzi E, Mantegazza C, Sartorio MUA, Pensabene L, Baldassarre ME, Agosti M, Vandenplas Y, Zuccotti G. Cow's Milk Allergy: Immunomodulation by Dietary Intervention. Nutrients 2019; 11:1399. [PMID: 31234330 PMCID: PMC6627562 DOI: 10.3390/nu11061399] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/26/2022] Open
Abstract
Cow's milk proteins cause allergic symptoms in 2% to 3% of all infants. In these individuals, the physiological mechanism of tolerance is broken with subsequent possible sensitization to antigens, which can lead eventually to allergic responses. The present review aims to provide an overview of different aspects of immune modulation by dietary intervention in cow's milk allergy (CMA). It focuses on pathogenetic mechanisms of different CMA related disorders, e.g., gastroesophageal reflux and eosinophilic esophagitis, highlighting the role of dietary management on innate and adaptive immune systems. The traditional dietary management of CMA has greatly changed in the last years, moving from a passive approach, consisting of an elimination diet to relieve symptoms, to a "proactive" one, meaning the possibility to actively modulate the immune system. Thus, new insights into the role of hydrolysates and baked milk in immunomodulation are addressed here. Additionally, nutritional components, such as pre- and probiotics, may target the immune system via microbiota, offering a possible road map for new CMA prevention and treatment strategies.
Collapse
Affiliation(s)
- Enza D'Auria
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Silvia Salvatore
- Department of Pediatrics, Ospedale "F. Del Ponte", University of Insubria, 21100 Varese, Italy.
| | - Elena Pozzi
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Cecilia Mantegazza
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Marco Ugo Andrea Sartorio
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| | - Licia Pensabene
- Department of Medical and Surgical Sciences, Pediatric Unit, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Maria Elisabetta Baldassarre
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Massimo Agosti
- Department of Pediatrics, Ospedale "F. Del Ponte", University of Insubria, 21100 Varese, Italy.
| | - Yvan Vandenplas
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| | - GianVincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children's Hospital-University of Milan, 20154 Milan, Italy.
| |
Collapse
|
30
|
Velasco Rodríguez-Belvís M, Viada Bris JF, Plata Fernández C, García-Salido A, Asensio Antón J, Domínguez Ortega G, Muñoz Codoceo RA. Normal fecal calprotectin levels in healthy children are higher than in adults and decrease with age. Paediatr Child Health 2019; 25:286-292. [PMID: 32765164 DOI: 10.1093/pch/pxz070] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
Background/Objectives The paediatric reference range of fecal calprotectin (FC) has not been decisively established and previous studies show a wide within-age variability, suggesting that other factors like anthropometric data or type of feeding can influence FC. Our aims were to establish the normal levels of FC in healthy children grouped by age and analyze whether sex, gestational age, birth weight, type of delivery, type of feeding, or anthropometric data influence FC values. Methods This multicentre, cross-sectional, and observational study enrolled healthy donors under 18 years of age who attended their Primary Health Care Centre for their routine Healthy Child Program visits. The exclusion criteria were: (i) immunodeficiency, (ii) autoimmune or (iii) gastrointestinal disease; (iv) medication usage; (v) gastrointestinal symptoms; or (vi) positive finding in the microbiological study. Results We enrolled 395 subjects, mean age was 4.2 years (range 3 days to 16.9 years), and 204 were male. The median FC was 77.0 mcg/g (interquartile range 246). A negative correlation between age and FC was observed (Spearman's rho = -0.603, P<0.01), and none of the other factors analyzed were found to influence FC levels. Conclusions Normal FC values in healthy children (particularly in infants) are higher than those considered to be altered in adults and show a negative correlation with age. It is necessary to reconsider the upper limits of FC levels for paediatric patients according to age, with further studies required to determine other factors that influence FC during infancy.
Collapse
Affiliation(s)
| | | | | | - Alberto García-Salido
- Pediatric Intensive Care Unit, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Julia Asensio Antón
- Clinical Analysis Department, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Gloria Domínguez Ortega
- Gastroenterology and Nutrition Department, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Rosa Ana Muñoz Codoceo
- Gastroenterology and Nutrition Department, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| |
Collapse
|
31
|
Abstract
Inflammatory bowel disease (IBD) denotes a group of chronic incurable disorders characterized by relapsing-remitting inflammation of the gastrointestinal tract. IBD represents a growing global burden with a prevalence exceeding 0.3% in the Western world and an accelerating incidence in newly industrialized countries. The target for treating IBD has shifted in recent years from symptom control to mucosal healing (MH), which has been shown to be associated with favorable long-term outcomes. The gold standard for ascertaining MH is endoscopic assessment, but endoscopy is limited by its invasive nature, high cost, and finite availability. Surrogate biomarkers are therefore of great utility. Calprotectin, a cytosolic protein derived predominantly from neutrophils, is now widely used in this capacity. Calprotectin is found in various bodily fluids at concentrations proportional to the degree of inflammation, including in feces at levels roughly six times higher than in the blood. Fecal calprotectin (FCP) therefore reflects intestinal inflammation. Various assays, including point-of-care and home-based tests, are now available for measuring FCP. FCP is used for screening purposes, to aid in distinguishing inflammatory from non-inflammatory gastrointestinal conditions like irritable bowel syndrome (IBS), as well as in the monitoring of known IBD. The aims of this review are to provide an overview of the methods used to measure FCP and to review the evidence supporting the use of FCP in IBD, particularly as it pertains to screening, monitoring and predicting disease relapse.
Collapse
Affiliation(s)
- Amanda Ricciuto
- a Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children , University of Toronto , Toronto , Canada
| | - Anne M Griffiths
- a Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children , University of Toronto , Toronto , Canada
| |
Collapse
|
32
|
Baldassarre ME, Di Mauro A, Capozza M, Rizzo V, Schettini F, Panza R, Laforgia N. Dysbiosis and Prematurity: Is There a Role for Probiotics? Nutrients 2019; 11:E1273. [PMID: 31195600 PMCID: PMC6627287 DOI: 10.3390/nu11061273] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/07/2019] [Accepted: 05/20/2019] [Indexed: 12/23/2022] Open
Abstract
Healthy microbiota is a critical mediator in maintaining health and it is supposed that dysbiosis could have a role in the pathogenesis of a number of diseases. Evidence supports the hypothesis that maternal dysbiosis could act as a trigger for preterm birth; aberrant colonization of preterm infant gut might have a role in feeding intolerance and pathogenesis of necrotizing enterocolitis. Despite several clinical trials and meta-analyses, it is still not clear if modulation of maternal and neonatal microbiota with probiotic supplementation decreases the risk of preterm birth and its complications.
Collapse
Affiliation(s)
- Maria Elisabetta Baldassarre
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Antonio Di Mauro
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Manuela Capozza
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Valentina Rizzo
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Federico Schettini
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Raffaella Panza
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| | - Nicola Laforgia
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, "Aldo Moro" University of Bari, P.zza Giulio Cesare 11, 70124 Bari, Italy.
| |
Collapse
|
33
|
Serial fecal calprotectin in the prediction of necrotizing enterocolitis in preterm neonates. J Pediatr Surg 2019; 54:455-459. [PMID: 29859621 DOI: 10.1016/j.jpedsurg.2018.04.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 11/21/2022]
Abstract
PURPOSE To investigate whether serial measurements of fecal calprotectin concentrations enable us to identify infants who will develop NEC prior to development of symptoms. METHODS Prospective matched case-control study including 100 high-risk neonates. High risk includes 1) gestational age (GA) ≤30 weeks, 2) birth-weight (BW) ≤1000 g, 3) GA 30-32 weeks and BW ≤1250 g, 4) born from a mother who received indomethacin for tocolysis. We matched every NEC subject with three controls for birth weight and gestational age. Fecal calprotectin was measured twice a week from day one until five weeks after birth or until NEC development. We analyzed differences in fecal calprotectin between NEC subjects and controls in the week preceding NEC onset and course of fecal calprotectin within subjects who developed NEC. RESULTS Of 100 included patients, ten (median GA 27.5 weeks [24.6-29.4], BW 1010 g [775-1630]) developed NEC. The median calprotectin concentration in all samples combined was 332 μg/g [<40-8230] μg/g feces. There were no differences between NEC subjects and controls, with a wide variation in both groups. In NEC subjects, there was no intraindividual rise in calprotectin before clinical symptoms occurred. CONCLUSIONS There are high concentrations and wide interindividual variations in calprotectin in preterm infants during the first weeks of life. Wide intraindividual variation further precludes the serial use of fecal calprotectin in the early detection or prediction of NEC in high risk infants. LEVEL OF EVIDENCE III.
Collapse
|
34
|
Henrick BM, Chew S, Casaburi G, Brown HK, Frese SA, Zhou Y, Underwood MA, Smilowitz JT. Colonization by B. infantis EVC001 modulates enteric inflammation in exclusively breastfed infants. Pediatr Res 2019; 86:749-757. [PMID: 31443102 PMCID: PMC6887859 DOI: 10.1038/s41390-019-0533-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/11/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Infant gut dysbiosis, often associated with low abundance of bifidobacteria, is linked to impaired immune development and inflammation-a risk factor for increased incidence of several childhood diseases. We investigated the impact of B. infantis EVC001 colonization on enteric inflammation in a subset of exclusively breastfed term infants from a larger clinical study. METHODS Stool samples (n = 120) were collected from infants randomly selected to receive either 1.8 × 1010 CFU B. infantis EVC001 daily for 21 days (EVC001) or breast milk alone (controls), starting at day 7 postnatal. The fecal microbiome was analyzed using 16S ribosomal RNA, proinflammatory cytokines using multiplexed immunoassay, and fecal calprotectin using ELISA at three time points: days 6 (Baseline), 40, and 60 postnatal. RESULTS Fecal calprotectin concentration negatively correlated with Bifidobacterium abundance (P < 0.0001; ρ = -0.72), and proinflammatory cytokines correlated with Clostridiaceae and Enterobacteriaceae, yet negatively correlated with Bifidobacteriaceae abundance. Proinflammatory cytokines were significantly lower in EVC001-fed infants on days 40 and 60 postnatally compared to baseline and compared to control infants. CONCLUSION Our findings indicate that gut dysbiosis (absence of B. infantis) is associated with increased intestinal inflammation. Early addition of EVC001 to diet represents a novel strategy to prevent enteric inflammation during a critical developmental phase.
Collapse
Affiliation(s)
- Bethany M. Henrick
- Evolve Biosystems, Inc, Davis, CA USA ,0000 0004 1937 0060grid.24434.35Department of Food Science and Technology, University of Nebraska, Lincoln, NE USA
| | | | | | | | - Steven A. Frese
- Evolve Biosystems, Inc, Davis, CA USA ,0000 0004 1937 0060grid.24434.35Department of Food Science and Technology, University of Nebraska, Lincoln, NE USA
| | - You Zhou
- 0000 0004 1937 0060grid.24434.35Morrison Microscopy Core Research Facility, University of Nebraska, Lincoln, NE USA
| | - Mark A. Underwood
- 0000 0004 1936 9684grid.27860.3bFoods for Health Institute, University of California Davis, Davis, CA USA ,0000 0004 1936 9684grid.27860.3bDepartment of Pediatrics, University of California Davis Children’s Hospital, Sacramento, CA USA
| | - Jennifer T. Smilowitz
- 0000 0004 1936 9684grid.27860.3bFoods for Health Institute, University of California Davis, Davis, CA USA ,0000 0004 1936 9684grid.27860.3bDepartment of Food Science and Technology, University of California Davis, Davis, CA USA
| |
Collapse
|
35
|
O'Connor DL, Kiss A, Tomlinson C, Bando N, Bayliss A, Campbell DM, Daneman A, Francis J, Kotsopoulos K, Shah PS, Vaz S, Williams B, Unger S. Nutrient enrichment of human milk with human and bovine milk-based fortifiers for infants born weighing <1250 g: a randomized clinical trial. Am J Clin Nutr 2018; 108:108-116. [PMID: 29878061 DOI: 10.1093/ajcn/nqy067] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/14/2018] [Indexed: 01/03/2023] Open
Abstract
Background Human milk-based fortifiers (HMBFs) are being adopted in neonatal care to enrich the nutrients in human milk for very low birth weight (VLBW) infants despite being costly and there being limited efficacy data. No randomized clinical trial has evaluated the use of HMBF compared with bovine milk-based fortifiers (BMBFs) in the absence of formula feeding. Objective To determine if HMBF compared with BMBF for routine nutrient enrichment of human milk improves feeding tolerance, reduces morbidity, reduces fecal calprotectin (a measure of gut inflammation), and supports the growth of infants <1250 g. Design In this blinded randomized clinical trial, infants born weighing <1250 g were recruited from neonatal units in Ontario, Canada between August 2014 and November 2015. The infants were fed mother's milk and donor milk as required. Fortification commenced at 100 mL/kg per day of HMBF (0.81 kcal/mL) or BMBF (0.72 kcal/mL) and advanced at 140 mL/kg per day to 0.88 and 0.78 kcal/mL, respectively. The primary outcome was percentage of infants with a feeding interruption for ≥12 h or a >50% reduction in feeding volume. Secondary outcomes included a dichotomous mortality and morbidity index (i.e., affirmative for any one of death, late-onset sepsis, necrotizing enterocolitis, chronic lung disease, or severe retinopathy of prematurity), fecal calprotectin, and growth. Results Of 232 eligible infants, 127 (54.7%) were randomized (n = 64 HMBF, n = 63 BMBF). Mean ± SD birth weight and gestational age of infants were 888 ± 201 g and 27.7 ± 2.5 wk, respectively. No statistically significant differences were identified in feeding interruptions [17/64 HMBF, 20/61 BMBF; unadjusted risk difference: -6.2% (95% CI: -22.2%, 9.8%)]. There was no statistically significant difference in the mortality and morbidity index (48.4% HMBF, 49.2% BMBF, adjusted P = 0.76), changes in fecal calprotectin, or growth z scores. Conclusions Among infants born weighing <1250 g and exclusively fed human milk, the use of HMBF did not improve feeding tolerance or reduce mortality and morbidity compared with BMBF. This trial was registered at clinicaltrials.gov as NCT02137473.
Collapse
Affiliation(s)
- Deborah L O'Connor
- Translational Medicine Program and Divisions of Neonatology and Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada.,Departments of Nutritional Sciences, Pediatrics, and Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,Department of Pediatrics, Sinai Health System, Toronto, Canada
| | - Alex Kiss
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada.,Evaluative and Clinical Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Christopher Tomlinson
- Translational Medicine Program and Divisions of Neonatology and Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada.,Translational Medicine Program and Divisions of Neonatology, The Hospital for Sick Children, Toronto, Canada.,Departments of Nutritional Sciences, Pediatrics, and Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,Departments of Pediatrics and Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Nicole Bando
- Translational Medicine Program and Divisions of Neonatology and Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada
| | - Ann Bayliss
- Trillium Health Partners, Mississauga, Canada
| | - Douglas M Campbell
- Translational Medicine Program and Divisions of Neonatology, The Hospital for Sick Children, Toronto, Canada.,Departments of Pediatrics and Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,St. Michael's Hospital and Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Alan Daneman
- Translational Medicine Program and Divisions of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada.,Departments of Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Jane Francis
- Translational Medicine Program and Divisions of Neonatology and Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada.,Departments of Nutritional Sciences, Pediatrics, and Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | | | - Prakesh S Shah
- Departments of Pediatrics and Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,Department of Pediatrics, Sinai Health System, Toronto, Canada
| | - Simone Vaz
- Department of Pediatrics, William Osler Health System, Brampton, Canada
| | - Brock Williams
- Translational Medicine Program and Divisions of Neonatology and Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada
| | - Sharon Unger
- Translational Medicine Program and Divisions of Neonatology, The Hospital for Sick Children, Toronto, Canada.,Departments of Pediatrics and Medical Imaging, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,Department of Pediatrics, Sinai Health System, Toronto, Canada
| | | |
Collapse
|
36
|
Jang HJ, Park JH, Kim CS, Lee SL, Lee WM. Amino Acid-Based Formula in Premature Infants with Feeding Intolerance: Comparison of Fecal Calprotectin Level. Pediatr Gastroenterol Hepatol Nutr 2018; 21:189-195. [PMID: 29992119 PMCID: PMC6037800 DOI: 10.5223/pghn.2018.21.3.189] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 12/01/2017] [Accepted: 12/29/2017] [Indexed: 01/13/2023] Open
Abstract
PURPOSE We investigated fecal calprotectin (FC) levels in preterm infants with and without feeding intolerance (FI), and compared the FC levels according to the type of feeding. METHODS The medical records of 67 premature infants were reviewed retrospectively. The fully enteral-fed infants were classified into two groups; the FI group (29 infants) and the control group (31 infants). Seven infants with necrotizing enterocolitis, sepsis, and perinatal asphyxia were excluded. If breast milk (BM) or preterm formula (PF) could not be tolerated by infants with FI, amino acid-based formula (AAF) was tried temporarily. Once FI improved, AAF was discontinued, and BM or PF was resumed. We investigated the FC levels according to the type of feeding. RESULTS Significant differences were found in gestational age, birth weight, age when full enteral feeding was achieved, and hospital stay between the FI and control group (p<0.05). The FC levels in the FI group were significantly higher than those in the control group (p<0.05). The FC levels in the AAF-fed infants with FI were significantly lower than those in the BM- or PF-fed infants (p<0.05). The growth velocities (g/d) and z scores were not significantly different between the FI and control group (p>0.05). CONCLUSION The FC levels in AAF-fed infants with FI showed significantly lower than those in the BM- or PF-fed infants with FI. The mitigation of gut inflammation through the decrease of FC levels in AAF-fed infants with FI could be presumed.
Collapse
Affiliation(s)
- Hyo-Jeong Jang
- Department of Pediatrics, Keimyung University School of Medicine, Daegu, Korea
| | - Jae Hyun Park
- Department of Pediatrics, Keimyung University School of Medicine, Daegu, Korea
| | - Chun Soo Kim
- Department of Pediatrics, Keimyung University School of Medicine, Daegu, Korea
| | - Sang Lak Lee
- Department of Pediatrics, Keimyung University School of Medicine, Daegu, Korea
| | - Won Mok Lee
- Department of Laboratory Medicine, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
37
|
De Andrés J, Manzano S, García C, Rodríguez JM, Espinosa-Martos I, Jiménez E. Modulatory effect of three probiotic strains on infants' gut microbial composition and immunological parameters on a placebo-controlled, double-blind, randomised study. Benef Microbes 2018; 9:573-584. [PMID: 29726280 DOI: 10.3920/bm2017.0132] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The gut microbiota plays a crucial role in gastrointestinal health. Current use of probiotics is aimed at modulating the bacterial gut composition to alleviate specific diseases. The safety and tolerance of three probiotic strains (Bifidobacterium longum subsp. infantis R0033, Lactobacillus helveticus R0052 and Bifidobacterium bifidum R0071) has recently been described. The objective of the present study was to analyse the microbiological composition and immunological parameters of faecal samples obtained from healthy infants from 3 to 12 months of age after receiving either one of the three probiotic strains or placebo for 8 weeks. 16S ribosomal RNA gene sequencing and multiplexing technology was used for analysis. Faecal sample analysis showed that the most abundant genus in all four groups of infants pre- and post-intervention was Bifidobacterium, representing approximately 50% of the sequences. After the intervention period the microbial composition of faecal samples in the probiotic groups did not display notable changes. In contrast, a decrease in different Bifidobacterium species, such as B. bifidum and Bifidobacterium breve and an increase in Bacteroides, Blautia, Clostridium, Coprococcus and Faecalibacterium genera was observed in the placebo group. The analysis of a wide range of immune factors in faecal samples suggests a modulatory effect by these three probiotic strains during the intervention period. The anti-inflammatory ratio interleukin (IL)-10/IL-12 increased at the end of the intervention period in the B. infantis R0033 group while the TNF-α/IL-10 ratio increased in the L. helveticus R0052 group. The decrease of the IL-10/IL-12 ratio together with the increase of the tumour necrosis factor alpha (TNF-α)/IL-10 ratio demonstrated a pro-inflammatory profile in the placebo group. In conclusion, the species profile of the microbiome observed in all three probiotic groups resembled that of a younger infant, similar to an unweaned infant, when compared to the placebo group which may also be related with an anti-inflammatory effect.
Collapse
Affiliation(s)
- J De Andrés
- 1 Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040Madrid, Spain
| | - S Manzano
- 1 Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040Madrid, Spain.,2 Probisearch S.L.U., C/Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| | - C García
- 2 Probisearch S.L.U., C/Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| | - J M Rodríguez
- 1 Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040Madrid, Spain
| | - I Espinosa-Martos
- 2 Probisearch S.L.U., C/Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| | - E Jiménez
- 1 Dpto. Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid, Av. Puerta de hierro s/n, 28040Madrid, Spain.,2 Probisearch S.L.U., C/Santiago Grisolía, 2, 28760 Tres Cantos, Spain
| |
Collapse
|
38
|
Nakayuenyongsuk W, Christofferson M, Stevenson DK, Sylvester K, Lee HC, Park KT. Point-of-Care Fecal Calprotectin Monitoring in Preterm Infants at Risk for Necrotizing Enterocolitis. J Pediatr 2018. [PMID: 29519542 DOI: 10.1016/j.jpeds.2017.12.069] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVE To establish baseline trends in fecal calprotectin, a protein excreted into the stool when there is neutrophilic inflammation in the bowel, in infants at risk for necrotizing enterocolitis (NEC). STUDY DESIGN We performed a prospective observational cohort study in infants with a birth weight of <1500 g without existing bowel disease at a level IV neonatal intensive care unit from October 2015 to September 2016. Stools were collected once daily for 30 days or until 32 weeks postmenstrual age and processed using the Fecal Calprotectin High Range Quantitative Quantum Blue assay. RESULTS In 64 preterm infants, during the first week after birth, 62% of infants had an initial stool sample with high baseline calprotectin levels (≥200 µg/g). In assessment of maternal and neonatal risk factors, maternal etiology for preterm birth (ie, eclamplsia or preeclampsia) was the only significant factor associated with high baseline calprotectin level. Two patients in the cohort developed NEC. Calprotectin levels for the entire cohort fluctuated during the observed period but generally increased in the third and fourth weeks after birth. CONCLUSIONS At-risk infants had highly variable fecal calprotectin levels, with maternal causes for preterm birth associated with higher baseline levels. More longitudinal data in infants with NEC are necessary to determine whether acute rises in fecal calprotectin levels prior to clinical diagnosis can be confirmed as a diagnostic or prognostic biomarker.
Collapse
Affiliation(s)
- Warapan Nakayuenyongsuk
- Department of Pediatrics, Division of Gastroenterology, Stanford University School of Medicine, Palo Alto, CA
| | - Megan Christofferson
- Department of Pediatrics, Division of Gastroenterology, Stanford University School of Medicine, Palo Alto, CA
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Karl Sylvester
- Departement of Surgery, Division of Pediatric Surgery, Stanford University School of Medicine, Palo Alto, CA
| | - Henry C Lee
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - K T Park
- Department of Pediatrics, Division of Gastroenterology, Stanford University School of Medicine, Palo Alto, CA.
| |
Collapse
|
39
|
Shores DR, Everett AD. Children as Biomarker Orphans: Progress in the Field of Pediatric Biomarkers. J Pediatr 2018; 193:14-20.e31. [PMID: 29031860 PMCID: PMC5794519 DOI: 10.1016/j.jpeds.2017.08.077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/04/2017] [Accepted: 08/30/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Darla R Shores
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD.
| | - Allen D Everett
- Division of Cardiology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
40
|
Abstract
Calprotectin is a 36kDa member of the S100 family of proteins. It is derived predominantly from neutrophils and has direct antimicrobial effects and a role within the innate immune response. Calprotectin is found in various body fluids in proportion to the degree of any existing inflammation and its concentration in feces is about six times that of plasma. Measurement of fecal calprotectin is a useful surrogate marker of gastrointestinal inflammation. It has a high negative predictive value in ruling out inflammatory bowel disease (IBD) in undiagnosed, symptomatic patients and a high sensitivity for diagnosing the disease making it useful as a tool for prioritising endoscopy. In patients with known IBD, fecal calprotectin can be a useful tool to assist management, providing evidence of relapse or mucosal healing to enable therapy to be intensified or reduced. There are a number of commercial calprotectin assays with marked difference in performance as judged by external quality assessment and at present no standardised reference material exists. Various factors may affect results including age, medication and day to day variation. Laboratories should therefore be mindful of the characteristics of their own assay and factors that may affect results.
Collapse
Affiliation(s)
- Ruth M Ayling
- FRCPath Consultant Chemical Pathologist, Clinical Biochemistry, Pathology and Pharmacy Building, Royal London Hospital, London, United Kingdom
| | - Klaartje Kok
- MRCP Consultant Gastroenterologist, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
41
|
Dewanto NE, Firmansyah A, Sungkar A, Dharmasetiawani N, Sastroasmoro S, Kresno SB, Suradi R, Bardosono S, Prasetyo D. The effect of <em>Bifidobacterium animalis lactis</em> HNO19 supplementation among pregnant and lactating women on interleukin-8 level in breast milk and infant’s gut mucosal integrity. MEDICAL JOURNAL OF INDONESIA 2017. [DOI: 10.13181/mji.v26i3.1481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Background: Newborn’s gut mucosal is not fully developed, therefore infants are prone to diarrhea. Probiotic supplementation is known to induce the gut mucosal maturity. This study aimed to identify whether probiotics supplementation among pregnant women since the third trimester would increase the infant’s gut mucosal integrity.Methods: A double-blind, randomized clinical trial was conducted to understand the potential effect of probiotic supplementation on the level of probiotics and IL-8 in breastmilk, urine IFABP, faecal α-1-antytripsin (AAT) and calprotectin in infant’s at birth (V0) and three-months old (V3). A single strain of Bifidobacterium lactis animalis HNO19 (known as DR10) was used since it was not the resident bacteria. The study was held at Budi Kemuliaan Hospital and its satellite clinics from December 2014 to December 2015.Results: About 14% (5/35) and 20% (7/35) of the subjects had DR10 in the breastmilk’s colostrum and at the age of 3-months. The median values of IL-8 in the probiotic group vs the placebo group at V0 and V3 were 2810,1 pg/mL vs 1516.4 pg/mL (p=0.327) and 173.2 pg/mL vs 132.7 pg/mL (p=0.211) respectively. IFABP level 211.7 ng/mL vs 842.5 ng/mL (p=0.243) and 25.3 ng/mL vs 25.1 ng/mL (p=0.466); AAT 136.2 mg/dL vs 148.1 mg/dL (p=0.466) and 24 mg/mL vs 29.72 mg/mL (p=0.545); Calprotectin 746.8 ng/mL vs 4645.2 ng/mL (p=0.233) and 378.6 ng/mL vs 391.3 ng/mL (p=0.888).Conclusion: Probiotic DR10 given to pregnant women since the 3rd trimester can be found in colostrum and 3-months breastmilk. However, it did not affect the level of other probiotics or IL-8 and the gut mucosal integrity.
Collapse
|
42
|
Ellemunter H, Schüller K, Steinkamp G. Faecal calprotectin concentrations in young children with cystic fibrosis. J Cyst Fibros 2017; 17:e8-e9. [PMID: 29107599 DOI: 10.1016/j.jcf.2017.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 12/01/2022]
Affiliation(s)
- Helmut Ellemunter
- Cystic Fibrosis (CF) Centre at the Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria.
| | | | - Gratiana Steinkamp
- Cystic Fibrosis (CF) Centre at the Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; Clinical Research and Medical Scientific Writing, Schwerin, Germany
| |
Collapse
|
43
|
Strunk T, Hibbert J, Doherty D, Granland C, Trend S, Simmer K, Burgner D, Patole S, Currie A. Probiotics and antimicrobial protein and peptide levels in preterm infants. Acta Paediatr 2017; 106:1747-1753. [PMID: 28294428 DOI: 10.1111/apa.13826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/22/2017] [Accepted: 03/07/2017] [Indexed: 12/26/2022]
Abstract
AIM To characterise the secreted and inducible antimicrobial protein and peptide (APP) levels in a prospective cohort of preterm infants (<30 weeks gestational age) with or without Bifidobacterium breve M16V supplementation during the first month of life. METHODS We analysed serial biosamples of infants who did (n = 13) or did not receive (n = 62) B. breve (3 × 109 cfu/day). Peripheral blood was obtained on days 1, 14 and 28, and infant stool prior to commencement of probiotic supplementation and on day 21. Levels of APP (bactericidal/permeability inducing protein (BPI), beta defensins 1 and 2, lactoferrin, human cathelicidin, secretory phospholipase A2) in plasma and stool were determined. Further, we characterised induced APP levels in whole blood cultured with live S. epidermidis or with agonists of Toll-like receptors 2/6 and 4. RESULTS Stool, plasma and stimulated blood APP levels changed significantly during the first month of life. Supplementation with B. breve did not affect basal or stimulated APP levels except for a transient increase in inducible BPI. CONCLUSION Supplementation with B. breve does not appear to act via modulation of systemic or enteric APP expression in preterm infants although small effects cannot be excluded. Further work with other probiotic preparations is warranted.
Collapse
Affiliation(s)
- Tobias Strunk
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- Neonatal Clinical Care Unit; King Edward Memorial Hospital; Perth WA Australia
| | - Julie Hibbert
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Dorota Doherty
- School of Women's and Infant's Health; University of Western Australia; Perth WA Australia
| | - Caitlyn Granland
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Stephanie Trend
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Karen Simmer
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- Neonatal Clinical Care Unit; King Edward Memorial Hospital; Perth WA Australia
| | - David Burgner
- Murdoch Childrens Research Institute; Royal Children's Hospital; Parkville VIC Australia
- Department of Paediatrics; University of Melbourne; Melbourne VIC Australia
| | - Sanjay Patole
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- Neonatal Clinical Care Unit; King Edward Memorial Hospital; Perth WA Australia
| | - Andrew Currie
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- School of Veterinary and Life Sciences; Murdoch University; Perth WA Australia
| |
Collapse
|
44
|
Georgountzou A, Papadopoulos NG. Postnatal Innate Immune Development: From Birth to Adulthood. Front Immunol 2017; 8:957. [PMID: 28848557 PMCID: PMC5554489 DOI: 10.3389/fimmu.2017.00957] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/26/2017] [Indexed: 12/20/2022] Open
Abstract
It is well established that adaptive immune responses are deficient in early life, contributing to increased mortality and morbidity. The developmental trajectories of different components of innate immunity are only recently being explored. Individual molecules, cells, or pathways of innate recognition and signaling, within different compartments/anatomical sites, demonstrate variable maturation patterns. Despite some discrepancies among published data, valuable information is emerging, showing that the developmental pattern of cytokine responses during early life is age and toll-like receptor specific, and may be modified by genetic and environmental factors. Interestingly, specific environmental exposures have been linked both to innate function modifications and the occurrence of chronic inflammatory disorders, such as respiratory allergies. As these conditions are on the rise, our knowledge on innate immune development and its modulating factors needs to be expanded. Improved understanding of the sequence of events associated with disease onset and persistence will lead toward meaningful interventions. This review describes the state-of-the-art on normal postnatal innate immune ontogeny and highlights research areas that are currently explored or should be further addressed.
Collapse
Affiliation(s)
- Anastasia Georgountzou
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G Papadopoulos
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Infection, Inflammation and Respiratory Medicine, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
45
|
Lee YM, Min CY, Choi YJ, Jeong SJ. Delivery and feeding mode affects fecal calprotectin levels in infants <7months old. Early Hum Dev 2017; 108:45-48. [PMID: 28391117 DOI: 10.1016/j.earlhumdev.2017.03.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Elevated fecal calprotectin (FC) is a non-invasive marker of inflammation indicating bowel disease. However, healthy infants have displayed high FC levels, with large variation. We sought to determine what factors might affect FC levels in children <6months old. METHODS We recruited 133 healthy infants aged 0-6months from four Korean day care centers. Stool samples were analyzed by immunosorbent assay. All infants completed a questionnaire related to clinical characteristics including birth and feeding history. RESULTS The mean FC concentration of all of participants was 237.40μg/g (range 11.5-1330.6μg/g). FC levels between 0 and 6months reduced with increasing age and the differences were statistically significant. The mean FC value according to feeding mode was 354.67μg/g for those fed breast milk and 149.44μg/g for those fed formula milk (p<0.001). Mean FC values were 319.69μg/g vs. 130.97μg/g for normal spontaneous vaginal delivery (NSVD) vs. caesarean section births, respectively (p<0.001). In addition, delivery mode affected the FC level at 0-6months regardless of feeding mode. CONCLUSION The FC levels of infants aged 0-6months reduced with age and were higher than the normal levels observed in healthy older infants. The FC value at <7months was higher in infants who were fed breast milk and born by NSVD.
Collapse
Affiliation(s)
- Yoo Mi Lee
- Department of Pediatrics, Bundang CHA Medical Center, CHA University, Seongnam, South Korea
| | - Chae-Yeon Min
- Department of Pediatrics, Bundang CHA Medical Center, CHA University, Seongnam, South Korea
| | - You Jin Choi
- Department of Pediatrics, Bundang CHA Medical Center, CHA University, Seongnam, South Korea
| | - Su Jin Jeong
- Department of Pediatrics, Bundang CHA Medical Center, CHA University, Seongnam, South Korea.
| |
Collapse
|
46
|
Normative Fecal Calprotectin Concentrations in Guatemalan Preschoolers Are High Relative to Children Reported Elsewhere. J Pediatr Gastroenterol Nutr 2017; 64:238-244. [PMID: 27101538 DOI: 10.1097/mpg.0000000000001241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Calprotectin is a fecal marker of intraintestinal inflammation derived from activated enteric neutrophils and macrophages. It is useful as a clinical marker in inflammatory bowel diseases; furthermore, it may have a role in public health epidemiology. OBJECTIVES The aim of the study was to describe the distribution of fecal calprotectin in Guatemalan preschool children sharing a common institutional diet; to relate it collectively to pediatric distributions in other geographic settings, and individually to concomitant indicators of intestinal infection or colonization and other descriptive features of the child. METHODS Fecal samples were collected in 87 subjects, ages 2 to 7 years across 3 daycare centers sharing a common institutional menu, but from different ecological settings. Stools were examined, variously by routine light microscopy, quantitative egg counts, and a Giardia antigen test, for microbiological diagnosis, and an ELISA assay for fecal calprotectin (CalproLab). RESULTS The median fecal calprotectin value was 58 mg/kg, with a mean of 98 ± 136 mg/kg and a range from 10 to 950 mg/kg; 61% of values were above the manufacturer's cut-off for elevated concentration and 51% exceeded an age-adjusted criterion. There were no associations between sex, age, growth indicators, or fecal microbiological findings by microscopy or ELISA assays, alone or in combination. The central tendency (mean or median) and distribution were generally shifted to the right in relation to comparable reports from children across the world literature. CONCLUSIONS Although specific, low-grade intestinal infections do not define calprotectin subgroups, right-shifted fecal calprotectin status in this population may reflect a general and diffuse stress of adverse environmental sanitation.
Collapse
|
47
|
Environmental Enteric Dysfunction Is Associated With Poor Linear Growth and Can Be Identified by Host Fecal mRNAs. J Pediatr Gastroenterol Nutr 2016; 63:453-459. [PMID: 27347722 PMCID: PMC5084633 DOI: 10.1097/mpg.0000000000001315] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Environmental enteric dysfunction (EED) can be assessed by the lactulose:mannitol (L:M) test. Our objective was to determine if selected host fecal transcripts were correlated with EED, and whether transcripts and clinical characteristics could be used to predict EED in rural African children. METHODS Demographic and sanitation characteristics, along with L:M testing and host fecal transcript analyses from 798 asymptomatic Malawian children aged 12 to 61 months were compared with linear growth over the subsequent 3 months. Fecal host mRNA analysis included quantification of expression of 18 transcripts associated with L:M. Permeability was categorized as normal (L:M ≤ 0.15), moderate (0.15<L:M<0.45) and severe (L:M ≥ 0.45), and random forest predictive models were created. RESULTS L:M was inversely correlated with linear growth over the subsequent 3 months (r = -0.32, P < 0.001) and severe EED was associated with stunting (P < 0.0001). Age younger than 24 months, weight-for-height z score <0, domesticated animals in the child's sleep environment, lack of a pit latrine combined with a potentially contaminated water source, and a recent history of diarrhea were associated with severe EED. A random forest model using CD53, HLA-DRA, MUC12, and TNF was 84% sensitive for severe EED and 83% sensitive for no EED. CONCLUSIONS Selected host fecal transcripts can be used in a random forest model as a noninvasive biomarker for categories of EED in rural African children.
Collapse
|
48
|
Abstract
To date, we encounter more and more pediatric patients with ulcerative colitis (UC). For yet unclear reasons, UC in pediatric patients seems to be a more aggressive and extensive disease than in their adult counterparts. In the majority of pediatric patients, the disease presents as pancolitis. The severity of the disease is reflected in the high use of corticosteroids and immunosuppressants and a high rate of surgery for medically refractory patients. The means by which to assess disease activity or to accurately predict its course are far from optimal. This review summarizes the current knowledge on the means for assessing UC activity in children. Research for developing new tools by which to monitor and forecast disease activity, are needed in all areas including invasive endoscopy, clinical evaluation, and treatment follow-up.
Collapse
Affiliation(s)
- Kaija-Leena Kolho
- a Children's Hospital , Helsinki University Central Hospital, University of Helsinki , Helsinki , Finland
| |
Collapse
|
49
|
Moussa R, Khashana A, Kamel N, Elsharqawy SE. Fecal calprotectin levels in preterm infants with and without feeding intolerance. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2016. [DOI: 10.1016/j.jpedp.2016.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
50
|
Moussa R, Khashana A, Kamel N, Elsharqawy SE. Fecal calprotectin levels in preterm infants with and without feeding intolerance. J Pediatr (Rio J) 2016; 92:486-92. [PMID: 27317868 DOI: 10.1016/j.jped.2015.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/24/2015] [Accepted: 11/25/2015] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVES To assess the level of fecal calprotectin in preterm neonates with feeding intolerance, as well as to evaluate it as a marker of feeding intolerance and to determine a cut-off level of fecal calprotectin in feeding intolerance. METHODS Analytical, multicenter, case-control study, which was carried out in neonatal intensive care units in Egypt, in a period from August 1, 2014 to March 1, 2015 on 52 preterm neonates. Neonates were classified into two groups; a study group including 26 neonates who met inclusion criteria and a control group including 26 neonates for comparison. RESULTS Fecal calprotectin levels ranged from 3.9μg/g to 971.8μg/g, and there was a significant increase in fecal calprotectin in the study group when compared to the control group (334.3±236.6μg/g vs. 42.0±38.2μg/g, respectively) with moderate inverse significant correlation between fecal calprotectin and birth weight. Furthermore, there was moderate, significant correlation between fecal calprotectin and duration of breastfeeding range. On the other hand, there was no correlation between fecal calprotectin and post-natal age, gestational age, or volume of feeding. A cut-off at the 67.0μg/g level, with 100.0% sensitivity and 76.9% specificity, was considered. CONCLUSION Fecal calprotectin level increased significantly in neonates with feeding intolerance; it can be used to detect early cases with necrotizing enterocolitis in neonates, but this subject still needs more investigations on more patients.
Collapse
Affiliation(s)
- Rehab Moussa
- Suez Canal University, Faculty of Medicine, Department of Pediatrics, Ismailia, Egypt
| | - Abdelmoneim Khashana
- Suez Canal University, Faculty of Medicine, Department of Pediatrics, Ismailia, Egypt; University of Oulu, Medical Research Center (MRC), Research Unit of Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology, Ophtalmology (PEDEGO), Oulu, Finland.
| | - Noha Kamel
- Suez Canal University, Faculty of Medicine, Department of Clinical Pathology, Ismailia, Egypt
| | | |
Collapse
|