51
|
Fatheree NY, Liu Y, Taylor CM, Hoang TK, Cai C, Rahbar MH, Hessabi M, Ferris M, McMurtry V, Wong C, Vu T, Dancsak T, Wang T, Gleason W, Bandla V, Navarro F, Tran DQ, Rhoads JM. Lactobacillus reuteri for Infants with Colic: A Double-Blind, Placebo-Controlled, Randomized Clinical Trial. J Pediatr 2017; 191:170-178.e2. [PMID: 28969890 PMCID: PMC6336100 DOI: 10.1016/j.jpeds.2017.07.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 07/07/2017] [Accepted: 07/19/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess the safety of probiotic Lactobacillus reuteri strain Deutsche Sammlung von Mikroorganismen (DSM) 17938 with daily administration to healthy infants with colic and to determine the effect of L reuteri strain DSM 17938 on crying, fussing, inflammatory, immune, and microbiome variables. STUDY DESIGN We performed a controlled, double-blinded, phase 1 safety and tolerability trial in healthy breast-fed infants with colic, aged 3 weeks to 3 months, randomly assigned to L reuteri strain DSM 17938 (5 × 108 colony-forming units daily) or placebo for 42 days and followed for 134 days. RESULTS Of 117 screened infants, 20 were randomized to L reuteri strain DSM 17938 or placebo (sunflower oil) (in a 2:1 ratio) with 80% retention. Eleven of the 20 (55%) presented with low absolute neutrophil counts (<1500/mm3), which resolved in all subjects by day 176. L reuteri strain DSM 17938 produced no severe adverse events and did not significantly change crying time, plasma bicarbonate, or inflammatory biomarkers. Fecal calprotectin decreased rapidly in both groups. In the infants with dominant fecal gram negatives (Klebsiella, Proteus, and Veillonella), resolution of colic was associated with marked decreases in these organisms. CONCLUSIONS Daily administration of L reuteri strain DSM 17938 appears to be safe in newborn infants with colic, including those with neutropenia, which frequently coexists. A placebo response of 66% suggests that many infants with colic will have resolution within 3 weeks. TRIAL REGISTRATION ClinicalTrials.gov: NCT01849991.
Collapse
Affiliation(s)
- Nicole Y Fatheree
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - Yuying Liu
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - Christopher M Taylor
- Department of Microbiology, Immunology & Parasitology Louisiana State University Health Sciences Center, New Orleans, LA
| | - Thomas K Hoang
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - Chunyan Cai
- Division of Clinical and Translational Sciences, Department of Internal Medicine, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX; Biostatistics/Epidemiology/Research Design (BERD) Component, Center for Clinical and Translational Sciences (CCTS), the University of Texas Health Science Center at Houston, Houston, TX
| | - Mohammad H Rahbar
- Division of Clinical and Translational Sciences, Department of Internal Medicine, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX; Biostatistics/Epidemiology/Research Design (BERD) Component, Center for Clinical and Translational Sciences (CCTS), the University of Texas Health Science Center at Houston, Houston, TX; Division of Epidemiology, Human Genetics, and Environmental Sciences (EHGES), University of Texas School of Public Health at Houston, TX
| | - Manouchehr Hessabi
- Biostatistics/Epidemiology/Research Design (BERD) Component, Center for Clinical and Translational Sciences (CCTS), the University of Texas Health Science Center at Houston, Houston, TX
| | - Michael Ferris
- Department of Microbiology, Immunology & Parasitology Louisiana State University Health Sciences Center, New Orleans, LA
| | - Valarie McMurtry
- Department of Microbiology, Immunology & Parasitology Louisiana State University Health Sciences Center, New Orleans, LA
| | - Christine Wong
- Memorial Hermann Hospital Investigational Drug Services, Memorial Hermann Hospital, Houston, TX
| | - Ta Vu
- Memorial Hermann Hospital Investigational Drug Services, Memorial Hermann Hospital, Houston, TX
| | - Theresa Dancsak
- Clinical Research Center, Memorial Hermann Hospital, Houston, TX
| | - Ting Wang
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - Wallace Gleason
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - Vinay Bandla
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - Fernando Navarro
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - Dat Q Tran
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX
| | - J Marc Rhoads
- Department of Pediatrics, the University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX.
| |
Collapse
|
52
|
Abstract
Necrotizing enterocolitis (NEC) is a multifactorial disease that occurs when multiple risk factors and/or stressors overlap, leading to profound inflammation and intestinal injury. Due to its multifactorial nature, there has been much uncertainty in identifying clear strategies for prevention of NEC. Despite these obstacles, the incidence of NEC has gradually been decreasing over the past 10 years, in part due to quality improvement (QI) initiatives to prevent NEC. Current QI strategies primarily target the various predisposing conditions. This article reviews the evidence on which QI interventions to prevent NEC have been based and provides examples of successful QI interventions.
Collapse
|
53
|
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in preterm infants and continues to be a major cause of morbidity and mortality. The incidence of NEC as well as mortality from the disease has persisted at unacceptably high levels for decades as current understanding of the cause remains incomplete. Identifying infants at risk and preventing NEC are mainstays of care. This article briefly examines disease presentation and treatment, identifies gaps in current understanding of disease pathology, and highlights new research that may lead to a decrease in the incidence of NEC in the future.
Collapse
|
54
|
Acid Suppressant Medications in the Neonatal Intensive Care Unit: Strategies for Appropriate Use. J Perinat Neonatal Nurs 2017; 31:299-302. [PMID: 29068849 DOI: 10.1097/jpn.0000000000000290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
55
|
He B, Hoang TK, Wang T, Ferris M, Taylor CM, Tian X, Luo M, Tran DQ, Zhou J, Tatevian N, Luo F, Molina JG, Blackburn MR, Gomez TH, Roos S, Rhoads JM, Liu Y. Resetting microbiota by Lactobacillus reuteri inhibits T reg deficiency-induced autoimmunity via adenosine A2A receptors. J Exp Med 2017; 214:107-123. [PMID: 27994068 PMCID: PMC5206500 DOI: 10.1084/jem.20160961] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/17/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
Regulatory T (T reg) cell deficiency causes lethal, CD4+ T cell-driven autoimmune diseases. Stem cell transplantation is used to treat these diseases, but this procedure is limited by the availability of a suitable donor. The intestinal microbiota drives host immune homeostasis by regulating the differentiation and expansion of T reg, Th1, and Th2 cells. It is currently unclear if T reg cell deficiency-mediated autoimmune disorders can be treated by targeting the enteric microbiota. Here, we demonstrate that Foxp3+ T reg cell deficiency results in gut microbial dysbiosis and autoimmunity over the lifespan of scurfy (SF) mouse. Remodeling microbiota with Lactobacillus reuteri prolonged survival and reduced multiorgan inflammation in SF mice. L. reuteri changed the metabolomic profile disrupted by T reg cell deficiency, and a major effect was to restore levels of the purine metabolite inosine. Feeding inosine itself prolonged life and inhibited multiorgan inflammation by reducing Th1/Th2 cells and their associated cytokines. Mechanistically, the inhibition of inosine on the differentiation of Th1 and Th2 cells in vitro depended on adenosine A2A receptors, which were also required for the efficacy of inosine and of L. reuteri in vivo. These results reveal that the microbiota-inosine-A2A receptor axis might represent a potential avenue for combatting autoimmune diseases mediated by T reg cell dysfunction.
Collapse
Affiliation(s)
- Baokun He
- Pediatrics Gastroenterology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
- Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Thomas K Hoang
- Pediatrics Gastroenterology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
- Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Ting Wang
- Pediatrics Gastroenterology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
- Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Michael Ferris
- Department of Microbiology, Immunology and Parasitology, Louisiana State University School of Medicine, New Orleans, LA 70118
| | - Christopher M Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University School of Medicine, New Orleans, LA 70118
| | - Xiangjun Tian
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University School of Medicine, New Orleans, LA 70118
| | - Dat Q Tran
- Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Jain Zhou
- Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Nina Tatevian
- Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Fayong Luo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Jose G Molina
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Michael R Blackburn
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Thomas H Gomez
- Center for Laboratory Animal Medicine and Care, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Stefan Roos
- Department of Microbiology, Uppsala BioCenter, Swedish University of Agricultural Sciences, 750 07 Uppsala, Sweden
- BioGaia AB, 103 64 Stockholm, Sweden
| | - J Marc Rhoads
- Pediatrics Gastroenterology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
- Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | - Yuying Liu
- Pediatrics Gastroenterology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
- Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| |
Collapse
|
56
|
Smith PB. Use of Reflux Medications in Premature Infants After Hospital Discharge. Pediatrics 2016; 138:e20162849. [PMID: 27940724 PMCID: PMC5127077 DOI: 10.1542/peds.2016-2849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2016] [Indexed: 11/24/2022] Open
Affiliation(s)
- P Brian Smith
- Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
57
|
Romaine A, Ye D, Ao Z, Fang F, Johnson O, Blake T, Benjamin DK, Cotten CM, Testoni D, Clark RH, Chu VH, Smith PB, Hornik CP. Safety of histamine-2 receptor blockers in hospitalized VLBW infants. Early Hum Dev 2016; 99:27-30. [PMID: 27390109 PMCID: PMC4969147 DOI: 10.1016/j.earlhumdev.2016.05.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/20/2016] [Accepted: 05/24/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Histamine-2 receptor (H2) blockers are often used in very low birth weight infants despite lack of population specific efficacy and safety data. AIMS We sought to describe safety and temporal trends in histamine-2 receptor (H2) blocker use in hospitalized very low birth weight (VLBW) infants. STUDY DESIGN We conducted a retrospective cohort study using a clinical database populated by an electronic health record shared by 348 neonatal intensive care units in the United States. SUBJECTS We included all VLBW infants without major congenital anomalies. OUTCOME MEASURES We used multivariable logistic regression with generalizing estimating equations to evaluate the association between days of H2 blocker exposure and risk of: 1) death or necrotizing enterocolitis (NEC); 2) death or sepsis; and 3) death, NEC, or sepsis. RESULTS Of 127,707 infants, 20,288 (16%) were exposed to H2 blockers for a total of 6,422,352days. Median gestational age for infants exposed to H2 blockers was 27weeks (25th 75th percentile 26, 29). H2 blocker use decreased from 18% of infants in 1997 to 8% in 2012 (p<0.001). On multivariable analysis, infants were at increased risk of the combined outcome of death, NEC, or sepsis on days exposed to H2 blockers (odds ratio=1.14) (95% confidence interval 1.08, 1.19). CONCLUSIONS H2 blocker use is associated with increased risk of the combined outcome of death, NEC, or sepsis in hospitalized VLBW infants.
Collapse
Affiliation(s)
| | - Daniel Ye
- Duke Clinical Research Institute, Durham, NC, USA.
| | - Zachary Ao
- Duke Clinical Research Institute, Durham, NC, USA.
| | - Francia Fang
- Duke Clinical Research Institute, Durham, NC, USA.
| | | | - Taylor Blake
- Duke Clinical Research Institute, Durham, NC, USA.
| | | | | | | | - Reese H Clark
- Pediatrix-Obstetrix Center for Research and Education, Sunrise, FL,USA.
| | - Vivian H Chu
- Duke Clinical Research Institute, Durham, NC, USA.
| | | | | |
Collapse
|
58
|
Vasylyeva TL, Singh R. Gut Microbiome and Kidney Disease in Pediatrics: Does Connection Exist? Front Microbiol 2016; 7:235. [PMID: 26973613 PMCID: PMC4776082 DOI: 10.3389/fmicb.2016.00235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/15/2016] [Indexed: 01/01/2023] Open
Abstract
Child development is a unique and continuous process that is impacted by genetics and environmental factors. Gut microbiome changes with development and depends on the stage of gut maturation, nutrition, and overall health. In spite of emerging data and active study in adults, the gut-renal axis in pediatrics has not been well considered and investigated. This review will focus on the current knowledge of gut microbiota impacts on kidney disease with extrapolation to the pediatric population.
Collapse
Affiliation(s)
- Tetyana L. Vasylyeva
- Department of Pediatrics, Texas Tech University Health Sciences Center, AmarilloTX, USA
| | | |
Collapse
|
59
|
The Developing Microbiome of the Preterm Infant. Clin Ther 2016; 38:733-9. [PMID: 26947798 DOI: 10.1016/j.clinthera.2016.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/03/2016] [Accepted: 02/03/2016] [Indexed: 12/26/2022]
Abstract
PURPOSE To determine the importance of the neonatal microbiome in intestinal and overall health. METHOD A review of existing literature. FINDINGS AND IMPLICATIONS The microbiome is increasingly understood to have a significant role in health and disease. However, the microbiome of the preterm infant is unique, with simple microbial communities exposed to a consistent diet in a regulated environment, and development from naive to stable under the influence of the neonatal intensive care unit. This early microbiome encounters a still developing host and thus has the potential to program fundamental pathways with implications for neonatal and later outcomes.
Collapse
|
60
|
Fatheree NY, Liu Y, Ferris M, Van Arsdall M, McMurtry V, Zozaya M, Cai C, Rahbar MH, Hessabi M, Vu T, Wong C, Min J, Tran DQ, Navarro F, Gleason W, Gonzalez S, Rhoads JM. Hypoallergenic formula with Lactobacillus rhamnosus GG for babies with colic: A pilot study of recruitment, retention, and fecal biomarkers. World J Gastrointest Pathophysiol 2016; 7:160-170. [PMID: 26909239 PMCID: PMC4753182 DOI: 10.4291/wjgp.v7.i1.160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 09/25/2015] [Accepted: 11/03/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate recruitment, retention, and estimates for effects of formula supplementation with Lactobacillus rhamnosus GG (LGG) on inflammatory biomarkers and fecal microbial community in infants with colic. METHODS A prospective, double-blind, placebo-controlled trial was conducted in otherwise healthy infants with colic. We screened 74 infants and randomized and analyzed results in 20 infants [9 receiving LGG (LGG+) and 11 not receiving LGG (LGG-)]. LGG was incorporated in the formula (Nutramigen(®)) (minimum of 3 × 10(7) CFU/d) in the LGG+ group. Fecal microbiota and inflammatory biomarkers, including fecal calprotectin (FC), plasma cytokines, circulating regulatory T cells (Tregs), and crying + fussing time were analyzed to determine optimal time points and effect sizes for a larger trial. RESULTS Recruitment in this population was slow, with about 66% of eligible infants willing to enroll; subject retention was better (75%). These rates were influenced by parents' reluctance to volunteer their infant for a clinical trial and by their tendency to change formulas. The maximal difference of crying + fussing time was observed at day 14, comparing the 2 groups, with a mean difference of -91 (95%CI: -76, 259) min (P = NS). FC showed no significant difference, but the optimal time to determine a potential effect was at day 90 [with a mean difference of 121 (95%CI: -48, 291) μg/g stool], observing a lower level of FC in the LGG+ group. The fecal microbial communities were chaotic, as determined by Shannon's diversity index and not apparently influenced by the probiotic. No significant change was observed in plasma inflammatory cytokines or Tregs, comparing LGG+ to LGG- groups. CONCLUSION Designing future colic trials involving a probiotic-supplemented formula for infants in the United States will require consideration for difficult enrollment. Infants with colic have major variations in feal microbiota and calprotectin, both of which improve with time, with optimal time points for measurement at days 14 and 90 after treatment.
Collapse
|
61
|
Vongbhavit K, Underwood MA. Prevention of Necrotizing Enterocolitis Through Manipulation of the Intestinal Microbiota of the Premature Infant. Clin Ther 2016; 38:716-32. [PMID: 26872618 DOI: 10.1016/j.clinthera.2016.01.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/30/2015] [Indexed: 12/17/2022]
Abstract
PURPOSE In spite of four decades of research, necrotizing enterocolitis (NEC) remains the most common gastrointestinal complication in premature infants with high mortality and long-term morbidity. The composition of the intestinal microbiota of the premature infant differs dramatically from that of the healthy term infant and appears to be an important risk factor for NEC. METHODS We review the evidence of an association between intestinal dysbiosis and NEC and summarize published English language clinical trials and cohort studies involving attempts to manipulate the intestinal microbiota in premature infants. FINDINGS Promising NEC prevention strategies that alter the intestinal microbiota include probiotics, prebiotics, synbiotics, lacteroferrin, and human milk feeding. IMPLICATIONS Shaping the intestinal microbiota of the premature infant through human milk feeding and dietary supplements decreases the risk of NEC. Further studies to identify the ideal microbial composition and the most effective combination of supplements are indicated.
Collapse
Affiliation(s)
- Kannikar Vongbhavit
- Department of Pediatrics, HRH Princess Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Nakornayok, Thailand; Department of Pediatrics, University of California Davis, Sacramento, California
| | - Mark A Underwood
- Department of Pediatrics, University of California Davis, Sacramento, California.
| |
Collapse
|
62
|
Abstract
OBJECTIVES The aim of the present study was to examine the changes in bacteria in hospitalized preterm infants during the first month of life. METHODS Rectal swabs were collected daily from 12 preterm infants. DNA was extracted from swabs from day of birth and weekly thereafter. Bacterial taxa were identified with next generation sequencing using universal bacterial primers targeted at the 16S ribosomal DNA on a 454 Roche titanium platform. Sequences were clustered into operational taxonomic units, and taxonomy was assigned against the Greengenes databank using Quantitative Insights Into Microbial Ecology version 1.4. Quantitative polymerase chain reaction was used to determine the abundance of Bifidobacterium spp. Functional assessment of the microbiome was performed with Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). RESULTS Average birth weight and gestational age were 1055 g and 28 weeks, respectively. There were 6 to 35 different bacterial families identified in the day-of-birth samples, unrelated to the mode of delivery. Richness decreased through hospitalization (week 1, 16.9 ± 7.7 vs weeks 3-5, 10.7 ± 3.4, P < 0.001). The Shannon diversity index demonstrated the lowest diversity at birth, an increase at week 2, followed by a rapid decline at weeks 3 to 5, suggesting the development of a more uniform microbiota composition after 2 weeks of stay at a neonatal intensive care unit. Enterobacteriaceae, Staphylococcaceae, and Enterococcaceae constituted the majority of the bacterial families. Bifidobacterium spp were infrequently detected at extremely low levels. PICRUSt analysis revealed the enhancement of peroxisome, PPAR, and adipocytokine signaling; plant-pathogen interaction; and aminobenzoate degradation pathways in week 1 samples. CONCLUSIONS Our results suggest that although preterm infants have individualized microbiota that are detectable at birth, the differences decrease during the neonatal intensive care unit hospitalization with increasing prominence of pathogenic microbiota.
Collapse
|
63
|
Abstract
The respiratory tract, once believed to be sterile, harbors diverse bacterial communities. The role of microorganisms within health and disease is slowly being unraveled. Evidence points to the neonatal period as a critical time for establishing stable bacterial communities and influencing immune responses important for long-term respiratory health. This review summarizes the evidence of early airway and lung bacterial colonization and the role the microbiome has on respiratory health in the short and long term. The challenges of neonatal respiratory microbiome studies and future research directions are also discussed.
Collapse
Affiliation(s)
- David J Gallacher
- Department of Child Health, School of Medicine, Cardiff University , Cardiff , UK
| | - Sailesh Kotecha
- Department of Child Health, School of Medicine, Cardiff University , Cardiff , UK
| |
Collapse
|
64
|
Abstract
Premature infants are at increased risk for morbidity and mortality due to necrotizing enterocolitis (NEC) and sepsis. Probiotics decrease the risk of NEC and death in premature infants; however, mechanisms of action are unclear. A wide variety of probiotic species have been evaluated for potential beneficial properties in vitro, in animal models, and in clinical trials of premature infants. Although there is variation by species and even strain, common mechanisms of protection include attenuation of intestinal inflammation, apoptosis, dysmotility, permeability, supplanting other gut microbes through production of bacteriocins, and more effective use of available nutrients. Here, we review the most promising probiotics and what is known about their impact on the innate and adaptive immune response.
Collapse
Affiliation(s)
- Mark A Underwood
- Chief Division of Neonatology, School of Medicine, University of California at Davis, Sacramento, CA
| |
Collapse
|
65
|
Histamine H2 Receptor-Mediated Suppression of Intestinal Inflammation by Probiotic Lactobacillus reuteri. mBio 2015; 6:e01358-15. [PMID: 26670383 PMCID: PMC4701830 DOI: 10.1128/mbio.01358-15] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Probiotics and commensal intestinal microbes suppress mammalian cytokine production and intestinal inflammation in various experimental model systems. Limited information exists regarding potential mechanisms of probiotic-mediated immunomodulation in vivo. In this report, we demonstrate that specific probiotic strains of Lactobacillus reuteri suppress intestinal inflammation in a trinitrobenzene sulfonic acid (TNBS)-induced mouse colitis model. Only strains that possess the hdc gene cluster, including the histidine decarboxylase and histidine-histamine antiporter genes, can suppress colitis and mucosal cytokine (interleukin-6 [IL-6] and IL-1β in the colon) gene expression. Suppression of acute colitis in mice was documented by diminished weight loss, colonic injury, serum amyloid A (SAA) protein concentrations, and reduced uptake of [18F]fluorodeoxyglucose ([18F]FDG) in the colon by positron emission tomography (PET). The ability of probiotic L. reuteri to suppress colitis depends on the presence of a bacterial histidine decarboxylase gene(s) in the intestinal microbiome, consumption of a histidine-containing diet, and signaling via the histamine H2 receptor (H2R). Collectively, luminal conversion of l-histidine to histamine by hdc+L. reuteri activates H2R, and H2R signaling results in suppression of acute inflammation within the mouse colon. Probiotics are microorganisms that when administered in adequate amounts confer beneficial effects on the host. Supplementation with probiotic strains was shown to suppress intestinal inflammation in patients with inflammatory bowel disease and in rodent colitis models. However, the mechanisms of probiosis are not clear. Our current studies suggest that supplementation with hdc+L. reuteri, which can convert l-histidine to histamine in the gut, resulted in suppression of colonic inflammation. These findings link luminal conversion of dietary components (amino acid metabolism) by gut microbes and probiotic-mediated suppression of colonic inflammation. The effective combination of diet, gut bacteria, and host receptor-mediated signaling may result in opportunities for therapeutic microbiology and provide clues for discovery and development of next-generation probiotics.
Collapse
|
66
|
Prescott HC, Dickson RP, Rogers MAM, Langa KM, Iwashyna TJ. Hospitalization Type and Subsequent Severe Sepsis. Am J Respir Crit Care Med 2015; 192:581-8. [PMID: 26016947 DOI: 10.1164/rccm.201503-0483oc] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Hospitalization is associated with microbiome perturbation (dysbiosis), and this perturbation is more severe in patients treated with antimicrobials. OBJECTIVES To evaluate whether hospitalizations known to be associated with periods of microbiome perturbation are associated with increased risk of severe sepsis after hospital discharge. METHODS We studied participants in the U.S. Health and Retirement Study with linked Medicare claims (1998-2010). We measured whether three hospitalization types associated with increasing severity of probable dysbiosis (non-infection-related hospitalization, infection-related hospitalization, and hospitalization with Clostridium difficile infection [CDI]) were associated with increasing risk for severe sepsis in the 90 days after hospital discharge. We used two study designs: the first was a longitudinal design with between-person comparisons and the second was a self-controlled case series design using within-person comparison. MEASUREMENTS AND MAIN RESULTS We identified 43,095 hospitalizations among 10,996 Health and Retirement Study-Medicare participants. In the 90 days following non-infection-related hospitalization, infection-related hospitalization, and hospitalization with CDI, adjusted probabilities of subsequent admission for severe sepsis were 4.1% (95% confidence interval [CI], 3.8-4.4%), 7.1% (95% CI, 6.6-7.6%), and 10.7% (95% CI, 7.7-13.8%), respectively. The incidence rate ratio (IRR) of severe sepsis was 3.3-fold greater during the 90 days after hospitalizations than during other observation periods. The IRR was 30% greater after an infection-related hospitalization versus a non-infection-related hospitalization. The IRR was 70% greater after a hospitalization with CDI than an infection-related hospitalization without CDI. CONCLUSIONS There is a strong dose-response relationship between events known to result in dysbiosis and subsequent severe sepsis hospitalization that is not present for rehospitalization for nonsepsis diagnoses.
Collapse
Affiliation(s)
- Hallie C Prescott
- 1 Department of Internal Medicine and.,2 Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan
| | | | - Mary A M Rogers
- 1 Department of Internal Medicine and.,2 Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan
| | - Kenneth M Langa
- 1 Department of Internal Medicine and.,2 Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan.,3 VA Center for Clinical Management Research, HSR&D Center of Innovation, Ann Arbor, Michigan; and.,4 Institute for Social Research, Ann Arbor, Michigan
| | - Theodore J Iwashyna
- 1 Department of Internal Medicine and.,2 Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan.,3 VA Center for Clinical Management Research, HSR&D Center of Innovation, Ann Arbor, Michigan; and.,4 Institute for Social Research, Ann Arbor, Michigan
| |
Collapse
|
67
|
Patel RM, Denning PW. Intestinal microbiota and its relationship with necrotizing enterocolitis. Pediatr Res 2015; 78:232-8. [PMID: 25992911 PMCID: PMC4655440 DOI: 10.1038/pr.2015.97] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 02/15/2015] [Indexed: 12/26/2022]
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in infants born prematurely. After birth, the neonatal gut must acquire a healthy complement of commensal bacteria. Disruption or delay of this critical process, leading to deficient or abnormal microbial colonization of the gut, has been implicated as key risk factor in the pathogenesis of NEC. Conversely, a beneficial complement of commensal intestinal microbiota may protect the immature gut from inflammation and injury. Interventions aimed at providing or restoring a healthy complement of commensal bacteria, such as probiotic therapy, are currently the most promising treatment to prevent NEC. Shifting the balance of intestinal microbiota from a pathogenic to protective complement of bacteria can protect the gut from inflammation and subsequent injury that leads to NEC. Herein, we review the relationship of intestinal microbiota and NEC in preterm infants.
Collapse
Affiliation(s)
- Ravi Mangal Patel
- 1] Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University School of Medicine, Atlanta, Georgia [2] Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Patricia W Denning
- 1] Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University School of Medicine, Atlanta, Georgia [2] Children's Healthcare of Atlanta, Atlanta, Georgia
| |
Collapse
|
68
|
Abstract
Bloodstream infections in the neonatal intensive care unit (NICU) are associated with many adverse outcomes in infants, including increased length of stay and cost, poor neurodevelopmental outcomes, and death. Attention to the insertion and maintenance of central lines, along with careful review of when the catheters can be safely discontinued, can minimize central-line-associated bloodstream infections rates. Good antibiotic stewardship can further decrease the incidence of bloodstream infections, minimize the emergence of drug-resistant organisms or Candida as pathogens in the NICU, and safeguard the use of currently available antibiotics for future infants.
Collapse
Affiliation(s)
- Joseph B Cantey
- Division of Neonatal/Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Division of Infectious Diseases, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| | - Aaron M Milstone
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, 200 North Wolfe Street, Room 3141, Baltimore, MD 21287, USA
| |
Collapse
|
69
|
Abstract
The field of genomics has expanded into subspecialties such as metagenomics over the course of the last decade and a half. The development of massively parallel sequencing capabilities has allowed for increasingly detailed study of the genome of the human microbiome, the microbial super organ that resides symbiotically within the mucosal tissues and integumentary system of the human host. The gut microbiome, and particularly the study of its origins in neonates, has become subtopics of great interest within the field of genomics. This brief review seeks to summarize recent literature regarding the origins and establishment of the neonatal gut microbiome, beginning in utero, and how it is affected by neonatal nutritional status (breastfed versus formula fed) and gestational age (term versus preterm). We also explore the role of dysbiosis, a perturbation within the fragile ecosystem of the microbiome, and its role in the origin of select pathologic states, specifically, obesity and necrotizing enterocolitis (NEC) in preterm infants. We discuss the evidence supporting enteral pre- and pro-biotic supplementation of commensal organisms such as Bifidobacterium and Lactobacillus in the neonatal period, and their role in the prevention and amelioration of NEC in premature infants. Finally, we review directions to consider for further research to promote human health within this field.
Collapse
Affiliation(s)
- Emily C Gritz
- Division of Perinatal Medicine, Department of Pediatrics, Yale Child Health Research Center, Yale University School of Medicine , New Haven, CT , USA
| | - Vineet Bhandari
- Division of Perinatal Medicine, Department of Pediatrics, Yale Child Health Research Center, Yale University School of Medicine , New Haven, CT , USA
| |
Collapse
|
70
|
McMurtry VE, Gupta RW, Tran L, Blanchard EE, Penn D, Taylor CM, Ferris MJ. Bacterial diversity and Clostridia abundance decrease with increasing severity of necrotizing enterocolitis. MICROBIOME 2015; 3:11. [PMID: 25810906 PMCID: PMC4373520 DOI: 10.1186/s40168-015-0075-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/24/2015] [Indexed: 05/21/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating neonatal gastrointestinal disease that primarily affects premature infants. It is characterized by bowel inflammation and necrosis. In spite of extensive research, there has been little progress in decreasing the incidence or mortality of NEC over the past three decades. The exact etiology of NEC has not been identified. However, it is believed to result from an inappropriate immune response to gut microbiota. Using 454-pyrosequencing analyses of 16S rRNA genes that were PCR-amplified from stool DNA specimens, we compared the gut microbiota of infants with NEC to matched controls without NEC. The infants with NEC were then categorized into three subgroups based on severity: mild, severe, and lethal. We compared the microbiota among these subgroups and between each severity group and appropriate controls. RESULTS Bacterial diversity and the relative abundance of Actinobacteria and Clostridia were significantly lower in NEC specimens compared to controls. The absence of Clostridia was significantly associated with NEC. Microbial diversity and Clostridia abundance and prevalence decreased with increasing severity of NEC. CONCLUSIONS Low bacterial diversity in stool specimens may be indicative of NEC and the severity of NEC. The low bacterial diversity, and the lack of Clostridia in lethal specimens, could indicate that the presence of a diverse bacterial population in the gut as well as the presence of taxa such as Clostridia may play a role in attenuating inflammation leading to NEC.
Collapse
Affiliation(s)
- Valarie E McMurtry
- />Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, 1901 Perdido St., New Orleans, Louisiana USA
| | - Raegan W Gupta
- />Department of Pediatrics, Children’s Hospital, 200 Henry Clay Ave., New Orleans, Louisiana USA
| | - Lynn Tran
- />Pediatrix Medical Group of Louisiana, Baton Rouge General Medical Center, Baton Rouge, LA USA
| | - Eugene E Blanchard
- />Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, 1901 Perdido St., New Orleans, Louisiana USA
| | - Duna Penn
- />Department of Pediatrics, Children’s Hospital, 200 Henry Clay Ave., New Orleans, Louisiana USA
| | - Christopher M Taylor
- />Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, 1901 Perdido St., New Orleans, Louisiana USA
| | - Michael J Ferris
- />Department of Pediatrics, Children’s Hospital, 200 Henry Clay Ave., New Orleans, Louisiana USA
- />The Research Institute for Children, 200 Henry Clay Ave., New Orleans, Louisiana USA
| |
Collapse
|
71
|
Abstract
Human milk is a complete source of nourishment for the infant. Exclusive breastfeeding not only sustains the infant's development but also guides the proliferation of a protective intestinal microbiota. Among the many components of milk that modulate the infant gut microbiota, the milk glycans, which comprise free oligosaccharides, glycoproteins, and glycolipids, are increasingly recognized as drivers of microbiota development and overall gut health. These glycans may display pleiotropic functions, conferring protection against infectious diseases and also acting as prebiotics, selecting for the growth of beneficial intestinal bacteria. The prebiotic effect of milk glycans has direct application to prevention of diseases such as necrotizing enterocolitis, a common and devastating disease of preterm infants. In this article, we review the impact of the human (and bovine) milk glycome on gut health through establishment of a milk-oriented microbiota in the neonate.
Collapse
Affiliation(s)
- Alline R. Pacheco
- Department of Viticulture and Enology, University of California, Davis, California 95616
- Foods for Health Institute, University of California, Davis, California 95616
| | - Daniela Barile
- Foods for Health Institute, University of California, Davis, California 95616
- Department of Food Science and Technology, University of California, Davis, California 95616
| | - Mark A. Underwood
- Foods for Health Institute, University of California, Davis, California 95616
- Department of Pediatrics, University of California, Davis, California 95616
| | - David A. Mills
- Department of Viticulture and Enology, University of California, Davis, California 95616
- Foods for Health Institute, University of California, Davis, California 95616
- Department of Food Science and Technology, University of California, Davis, California 95616
| |
Collapse
|
72
|
Underwood MA, Arriola J, Gerber CW, Kaveti A, Kalanetra KM, Kananurak A, Bevins CL, Mills DA, Dvorak B. Bifidobacterium longum subsp. infantis in experimental necrotizing enterocolitis: alterations in inflammation, innate immune response, and the microbiota. Pediatr Res 2014; 76:326-33. [PMID: 25000347 PMCID: PMC4167942 DOI: 10.1038/pr.2014.102] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 04/08/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Probiotics decrease the risk of necrotizing enterocolitis (NEC). We sought to determine the impact of Bifidobacterium longum subsp. infantis (B. infantis) in the established rat model of NEC. METHODS Rat pups delivered 1 d prior to term gestation were assigned to one of three groups: dam fed (DF), formula fed (FF), or fed with formula supplemented with 5 × 10(6) CFU B. infantis per day (FF+Binf). Experimental pups were exposed to hypoxia and cold stress. Ileal tissue was examined for pathology and expression of inflammatory mediators, antimicrobial peptides, and goblet-cell products. Ceca were assessed for bacterial composition by analysis of the 16S rRNA sequence. RESULTS Administration of B. infantis significantly reduced the incidence of NEC, decreased expression of Il6, Cxcl1, Tnfa, Il23, and iNOS, and decreased expression of the antimicrobial peptides Reg3b and Reg3g. There was significant microbial heterogeneity both within groups and between experiments. The cecal microbiota was not significantly different between the FF and FF+Binf groups. Bifidobacteria were not detected in the cecum in significant numbers. CONCLUSION In the rat model, the inflammation associated with NEC was attenuated by administration of probiotic B. infantis. Dysbiosis was highly variable, precluding determination of the precise role of the microbiota in experimental NEC.
Collapse
Affiliation(s)
- Mark A. Underwood
- Department of Pediatrics, University of California Davis, Sacramento, CA,Corresponding author: Mark A. Underwood MD; Division of Neonatology, 2516 Stockton Blvd, Sacramento, CA 95817, USA;
| | - Jennifer Arriola
- Department of Pediatrics and the Steele Children’s Research Center, The University of Arizona, Tucson, AZ
| | - Colin W. Gerber
- Department of Pediatrics and the Steele Children’s Research Center, The University of Arizona, Tucson, AZ
| | - Ashwini Kaveti
- Department of Pediatrics and the Steele Children’s Research Center, The University of Arizona, Tucson, AZ
| | - Karen M. Kalanetra
- Department of Viticulture and Enology, University of California Davis, Davis CA
| | - Anchasa Kananurak
- Department of Microbiology and Immunology, University of California Davis, Davis, CA
| | - Charles L. Bevins
- Department of Microbiology and Immunology, University of California Davis, Davis, CA
| | - David A. Mills
- Department of Viticulture and Enology, University of California Davis, Davis CA
| | - Bohuslav Dvorak
- Department of Pediatrics and the Steele Children’s Research Center, The University of Arizona, Tucson, AZ
| |
Collapse
|
73
|
|
74
|
Chandrasekaran M, Fleming P. Question 1: does the use of ranitidine increase the risk of NEC in preterm infants? Arch Dis Child 2014; 99:390-2. [PMID: 24626320 DOI: 10.1136/archdischild-2013-304973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
75
|
|
76
|
Abstract
Necrotizing enterocolitis (NEC) continues to be the most severe gastrointestinal emergency facing the preterm neonate. The pathogenesis of NEC is still a complex and poorly understood process, but with increasing understanding of the role of enteral feeding, gut immunity and the altered gut microbiota, new opportunities to reduce overall NEC rates are now possible. Prevention strategies continue to lead as the most suitable approaches to reducing NEC, as early diagnosis and rapid effective treatment of NEC are still not optimal. Programmatic changes are equally important as subscribing to individual prevention strategies. The primary focus of this review is to summarize the best strategies we currently have to eliminate NEC within an institution.
Collapse
|
77
|
Ellis CL, Bokulich NA, Kalanetra KM, Mirmiran M, Elumalai J, Haapanen L, Schegg T, Rutledge JC, Raff G, Mills DA, Underwood MA. Probiotic administration in congenital heart disease: a pilot study. J Perinatol 2013; 33:691-7. [PMID: 23599119 PMCID: PMC3758394 DOI: 10.1038/jp.2013.41] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 03/18/2013] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the impact of probiotic Bifidobacterium longum ssp. infantis on the fecal microbiota and plasma cytokines in neonates with congenital heart disease. STUDY DESIGN Sixteen infants with congenital heart disease were randomly assigned to receive either B. infantis (4.2 × 10(9) colony-forming units two times daily) or placebo for 8 weeks. Stool specimens from enrolled infants and from six term infants without heart disease were analyzed for microbial composition. Plasma cytokines were analyzed weekly in the infants with heart disease. RESULTS Healthy control infants had increased total bacteria, total Bacteroidetes and total bifidobacteria compared to the infants with heart disease, but there were no significant differences between the placebo and probiotic groups. Plasma interleukin (IL)10, interferon (IFN)γ and IL1β levels were transiently higher in the probiotic group. CONCLUSION Congenital heart disease in infants is associated with dysbiosis. Probiotic B. infantis did not significantly alter the fecal microbiota. Alterations in plasma cytokines were found to be inconsistent.
Collapse
|
78
|
Corvaglia L, Monari C, Martini S, Aceti A, Faldella G. Pharmacological therapy of gastroesophageal reflux in preterm infants. Gastroenterol Res Pract 2013; 2013:714564. [PMID: 23878533 PMCID: PMC3710644 DOI: 10.1155/2013/714564] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 05/22/2013] [Accepted: 06/12/2013] [Indexed: 02/07/2023] Open
Abstract
Although gastroesophageal reflux (GER) is a very common phenomenon among preterm infants, its therapeutic management is still an issue of debate among neonatologists. A step-wise approach should be advisable, firstly promoting nonpharmacological interventions and limiting drugs to selected infants unresponsive to the conservative measures or who are suffering from severe GER with clinical complications. Despite of this, a concerning pharmacological overtreatment has been increasingly reported. Most of the antireflux drugs, however, have not been specifically assessed in preterm infants; moreover, serious adverse effects have been noticed in association to their administration. This review mainly aims to draw the state of the art regarding the pharmacological management of GER in preterm infants, analyzing the best piecies of evidence currently available on the most prescribed anti-reflux drugs. Although further trials are required, sodium alginate-based formulations might be considered promising; however, data regarding their safety are still limited. Few piecies of evidence on the efficacy of histamine-2 receptor blockers and proton pump inhibitors in preterm infants with GER are currently available. Nevertheless, a significantly increased risk of necrotizing enterocolitis and infections has been largely reported in association with their use, thereby leading to an unfavorable risk-benefit ratio. The efficacy of metoclopramide in GER's improvement still needs to be clarified. Other prokinetic agents, such as domperidone and erythromycin, have been reported to be ineffective, whereas cisapride has been withdrawn due to its remarkable cardiac adverse effects.
Collapse
Affiliation(s)
- Luigi Corvaglia
- Neonatology and Neonatal Intensive Care Unit, S. Orsola-Malpighi Hospital, via Massarenti 11, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Caterina Monari
- Neonatology and Neonatal Intensive Care Unit, S. Orsola-Malpighi Hospital, via Massarenti 11, 40138 Bologna, Italy
| | - Silvia Martini
- Neonatology and Neonatal Intensive Care Unit, S. Orsola-Malpighi Hospital, via Massarenti 11, 40138 Bologna, Italy
| | - Arianna Aceti
- Neonatology and Neonatal Intensive Care Unit, S. Orsola-Malpighi Hospital, via Massarenti 11, 40138 Bologna, Italy
| | - Giacomo Faldella
- Neonatology and Neonatal Intensive Care Unit, S. Orsola-Malpighi Hospital, via Massarenti 11, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| |
Collapse
|