Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Nature ; 591(7851): 633-638, 2021 03.
Article in English | MEDLINE | ID: mdl-33627867

ABSTRACT

The gut microbiota of preterm infants develops predictably1-7, with pioneer species colonizing the gut after birth, followed by an ordered succession of microorganisms. The gut microbiota is vital to the health of preterm infants8,9, but the forces that shape these predictable dynamics of microbiome assembly are unknown. The environment, the host and interactions between microorganisms all potentially shape the dynamics of the microbiota, but in such a complex ecosystem, identifying the specific role of any individual factor is challenging10-14. Here we use multi-kingdom absolute abundance quantification, ecological modelling and experimental validation to address this challenge. We quantify the absolute dynamics of bacteria, fungi and archaea in a longitudinal cohort of 178 preterm infants. We uncover microbial blooms and extinctions, and show that there is an inverse correlation between bacterial and fungal loads in the infant gut. We infer computationally and demonstrate experimentally in vitro and in vivo that predictable assembly dynamics may be driven by directed, context-dependent interactions between specific microorganisms. Mirroring the dynamics of macroscopic ecosystems15-17, a late-arriving member of the microbiome, Klebsiella, exploits the pioneer microorganism, Staphylococcus, to gain a foothold within the gut. Notably, we find that interactions between different kingdoms can influence assembly, with a single fungal species-Candida albicans-inhibiting multiple dominant genera of gut bacteria. Our work reveals the centrality of simple microbe-microbe interactions in shaping host-associated microbiota, which is critical both for our understanding of microbiota ecology and for targeted microbiota interventions.


Subject(s)
Biodiversity , Gastrointestinal Microbiome , Infant, Premature , Bacterial Load , Diet , Female , Gestational Age , Humans , Infant , Infant, Newborn , Male , Microbial Interactions , Reproducibility of Results
2.
Pediatr Res ; 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39069538

ABSTRACT

BACKGROUND: Regional fat distribution may be a marker of metabolic health and brain growth in preterm infants. Point of care ultrasound has been used to assess regional fat in term infants but has not been used widely in preterm infants. OBJECTIVE: To longitudinally quantify changes in body composition metrics using bedside ultrasound in very preterm infants. STUDY DESIGN: Very preterm infants (N = 69) were enrolled after birth and body composition assessments were done through 36 completed weeks' postmenstrual age (PMA). Linear mixed effects regression was used to model change in body composition assessments over time. RESULTS: There was an average increase across PMA for each body composition outcome. Biceps ultrasound subcutaneous fat (SQF) thickness increased by 0.11 mm (95% CI: 0.09, 0.13) each postmenstrual week. Triceps, subscapular, and abdominal ultrasound SQF remained constant through 28 weeks' PMA, then increased each week through 36 completed weeks' PMA. The inter-rater and intra-rater intraclass correlation coefficients for the ultrasound SQF measures ranged from 85.8 to 99.9. CONCLUSION: Use of ultrasound as a novel method to assess regional fat distribution in very preterm infants is feasible and reliable. IMPACT: Regional fat distribution may be a marker of metabolic health and brain growth in preterm infants. Gold standard body composition assessments may not be feasible in medically fragile very preterm infants. This study assesses longitudinally changes in regional adiposity development using bedside ultrasound techniques in a multicenter cohort of very preterm infants. Results of this study show that bedside ultrasound as a novel method to assess regional subcutaneous fat distribution and development in very preterm infants is both feasible and reliable.

3.
J Pediatr ; 270: 113774, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37839510

ABSTRACT

OBJECTIVE: To determine if oral secretions (OS) can be used as a noninvasively collected body fluid, in lieu of tracheal aspirates (TA), to track respiratory status and predict bronchopulmonary dysplasia (BPD) development in infants born <32 weeks. STUDY DESIGN: This was a retrospective, single center cohort study that included data and convenience samples from week-of-life (WoL) 3 from 2 independent preterm infant cohorts. Using previously banked samples, we applied our sample-sparing, high-throughput proteomics technology to compare OS and TA proteomes in infants born <32 weeks admitted to the Neonatal Intensive Care Unit (NICU) (Cohort 1; n = 23 infants). In a separate similar cohort, we mapped the BPD-associated changes in the OS proteome (Cohort 2; n = 17 infants including 8 with BPD). RESULTS: In samples collected during the first month of life, we identified 607 proteins unique to OS, 327 proteins unique to TA, and 687 overlapping proteins belonging to pathways involved in immune effector processes, neutrophil degranulation, leukocyte mediated immunity, and metabolic processes. Furthermore, we identified 37 OS proteins that showed significantly differential abundance between BPD cases and controls: 13 were associated with metabolic and immune dysregulation, 10 of which (eg, SERPINC1, CSTA, BPI) have been linked to BPD or other prematurity-related lung disease based on blood or TA investigations, but not OS. CONCLUSIONS: OS are a noninvasive, easily accessible alternative to TA and amenable to high-throughput proteomic analysis in preterm newborns. OS samples hold promise to yield actionable biomarkers of BPD development, particularly for prospective categorization and timely tailored treatment of at-risk infants with novel therapies.

4.
J Pediatr Gastroenterol Nutr ; 76(2): 206-212, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36705701

ABSTRACT

OBJECTIVES: Preterm infants are born functionally pancreatic insufficient with decreased pancreatic production of lipase and proteases. Developmental pancreatic insufficiency (PI) may contribute to reduced nutrient absorption and growth failure. We sought to determine longitudinal fecal elastase (ELA1) levels in a cohort of preterm infants and whether levels are associated with growth outcomes. METHODS: Prospective observational study of 30 infants 24-34 weeks gestational age and birth weight ≤1250 g fed the exclusive human milk diet, consisting of human milk with human milk-based fortifier. ELA1 was quantified by ELISA during the first 2 weeks of life [Early; 7.5 ± 1.8 days of life (DOL)] and after attainment of full, fortified feedings (Late; 63.6 ± 24.1 DOL). RESULTS: Early ELA1 levels were 192.2 ± 96.4 µg/g, and Late ELA1 levels were 268.0 ± 80.3 µg/g, 39.4% higher (P = 0.01). Infants with early PI (ELA1 < 200 µg/g) were more likely male and of lower gestational age, weight, length, and head circumference at birth. These variables, but not PI status, independently predicted somatic growth. CONCLUSIONS: Fecal ELA1 in preterm infants fed exclusive human milk diet increases with postnatal age. Although pancreatic function in preterm infants may serve as a biological contributor to early postnatal growth failure, additional studies using fecal ELA1 as a predictive biomarker for growth failure are needed in larger cohorts.


Subject(s)
Food, Fortified , Infant, Premature , Infant , Infant, Newborn , Male , Humans , Weight Gain , Milk, Human , Pancreatic Elastase , Infant Nutritional Physiological Phenomena
5.
J Pediatr ; 244: 224-229.e3, 2022 05.
Article in English | MEDLINE | ID: mdl-35093319

ABSTRACT

This retrospective cohort study sought to identify the association between certain xenobiotic metabolites in maternal breast milk and the diagnoses of bronchopulmonary dysplasia and retinopathy of prematurity in extremely preterm infants. Several acetaminophen metabolites were associated with a 3- to 6-fold increased odds of these disorders, and metabolites of certain food products, benzoate, and caffeine were associated with decreased odds.


Subject(s)
Bronchopulmonary Dysplasia , Infant, Premature, Diseases , Retinopathy of Prematurity , Acetaminophen/adverse effects , Cohort Studies , Female , Humans , Infant , Infant, Extremely Premature , Infant, Newborn , Milk, Human , Retinopathy of Prematurity/diagnosis , Retrospective Studies , Xenobiotics
6.
J Pediatr ; 240: 37-43.e1, 2022 01.
Article in English | MEDLINE | ID: mdl-34508750

ABSTRACT

OBJECTIVE: To examine associations of systemic inflammation with growth outcomes at neonatal intensive care unit discharge or transfer among infants with extremely low gestational ages. STUDY DESIGN: We studied 850 infants at born at 23-27 weeks of gestation. We defined inflammatory protein elevation as the highest quartile of C-reactive protein (CRP), Interleukin (IL)-6, tumor necrosis factor-∝, or IL-8 on postnatal days 1, 7, and 14. We compared z-scores of weight, length, and head circumference at neonatal intensive care unit discharge or transfer between infants with vs without inflammatory protein elevation, adjusting in linear regression for birth size z-score, sex, gestational age, diet, comorbidities, medications, and length of hospitalization. RESULTS: The mean gestational age was 25 weeks (range, 23-27 weeks) and birth weight z-score 0.14 (range, -2.73 to 3.28). Infants with a high CRP on day 7 had lower weights at discharge or transfer (-0.17 z-score; 95% CI, -0.27 to -0.06) than infants without CRP elevation, with similar results on day 14. Infants with CRP elevation on day 14 were also shorter (-0.21 length z-scores; 95% CI, -0.38 to -0.04), and had smaller head circumferences (-0.18 z-scores; 95% CI, -0.33 to -0.04) at discharge or transfer. IL-6 elevation on day 14 was associated with lower weight (-0.12; 95% CI, -0.22 to -0.02); IL-6 elevation on day 7 was associated with shorter length (-0.27; 95% CI, -0.43 to -0.12). Tumor necrosis factor-∝ and IL-8 elevation on day 14 were associated with a lower weight at discharge or transfer. CONCLUSIONS: Postnatal systemic inflammation may contribute to impaired nutrient accretion during a critical period in development in infants with extremely low gestational ages.


Subject(s)
Infant, Extremely Premature/growth & development , Inflammation/physiopathology , Biomarkers , Body Height , Body Weight , C-Reactive Protein/analysis , Cephalometry , Female , Gestational Age , Hospitalization , Humans , Infant, Extremely Premature/physiology , Infant, Newborn , Inflammation/blood , Intensive Care Units, Neonatal , Interleukin-6/blood , Interleukin-8/blood , Male , Tumor Necrosis Factor-alpha/blood
7.
Transfusion ; 62(6): 1177-1187, 2022 06.
Article in English | MEDLINE | ID: mdl-35522536

ABSTRACT

BACKGROUND: Platelet transfusions (PTxs) are often given to septic preterm neonates at high platelet count thresholds in an attempt to reduce bleeding risk. However, the largest randomized controlled trial (RCT) of neonatal transfusion thresholds found higher mortality and/or major bleeding in infants transfused at higher thresholds. Using a murine model, we investigated the effects of adult PTx on neonatal sepsis-induced mortality, systemic inflammation, and platelet consumption. STUDY DESIGN AND METHODS: Polymicrobial sepsis was induced via intraperitoneal injection of cecal slurry preparations (CS1, 2, 3) into P10 pups. Two hours after infection, pups were transfused with washed adult Green Flourescent Protein (GFP+) platelets or control. Weights, platelet counts, and GFP% were measured before 4 and 24 h post-infection. At 24 h, blood was collected for quantification of plasma cytokines. RESULTS: The CS batches varied in 24 h mortality (11%, 73%, and 30% in CS1, 2, and 3, respectively), due to differences in bacterial composition. PTx had differential effects on sepsis-induced mortality and systemic inflammatory cytokines, increasing both in mice infected with CS1 (low mortality) and decreasing both in mice infected with CS2 and 3. In a mathematical model of platelet kinetics, the consumption of transfused adult platelets was higher than that of endogenous neonatal platelets, regardless of CS batch. DISCUSSION: Our findings support the hypothesis that transfused adult platelets are consumed faster than endogenous neonatal platelets in sepsis and demonstrate that PTx can enhance or attenuate neonatal inflammation and mortality in a model of murine polymicrobial sepsis, depending on the composition of the inoculum and/or the severity of sepsis.


Subject(s)
Neonatal Sepsis , Sepsis , Animals , Cytokines , Disease Models, Animal , Humans , Mice , Neonatal Sepsis/therapy , Platelet Transfusion , Sepsis/therapy
8.
Am J Physiol Gastrointest Liver Physiol ; 320(2): G227-G239, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33236951

ABSTRACT

Necrotizing enterocolitis (NEC) is a manifestation of maladaptive intestinal responses in preterm infants centrally medicated by unattenuated inflammation. Early in the postnatal period, preterm infants develop a deficit in arachidonic and docosahexaenoic acid, both potent regulators of inflammation. We hypothesized that the fatty acid composition of parenteral lipid emulsions uniquely induces blood and intestinal fatty acid profiles which, in turn, modifies the risk of NEC development. Forty-two preterm pigs were randomized to receive one of three lipid emulsions containing 100% soybean oil (SO), 15% fish oil (MO15), or 100% fish oil (FO100) with enteral feedings over an 8-day protocol. Blood and distal ileum tissue were collected for fatty acid analysis. The distal ileum underwent histologic, proteomic, and metabolomic analyses. Eight pigs [3/14 SO (21%), 3/14 MO15 (21%), and 2/14 FO100 (14%)] developed NEC. No differences in NEC risk were evident between groups despite differences in induced fatty acid profiles in blood and ileal tissue. Metabolomic analysis of NEC versus no NEC tissue revealed differences in tryptophan metabolism and arachidonic acid-containing glycerophospholipids. Proteomic analysis demonstrated no differences by lipid group; however, 15 proteins differentiated NEC versus no NEC in the domains of tissue injury, glucose uptake, and chemokine signaling. Exposure to parenteral lipid emulsions induces unique intestinal fatty acid and metabolomic profiles; however, these profiles are not linked to a difference in NEC development. Metabolomic and proteomic analyses of NEC versus no NEC intestinal tissue provide mechanistic insights into the pathogenesis of NEC in preterm infants.NEW & NOTEWORTHY Exposure to parenteral lipid emulsions induces unique intestinal fatty acid and metabolomic profiles; however, these profiles are not linked to a difference in NEC risk in preterm pigs. Metabolomic and proteomic analyses provide mechanistic insights into NEC pathogenesis. Compared with healthy ileal tissue, metabolites in tryptophan metabolism and arachidonic acid-containing glycerophospholipids are increased in NEC tissue. Proteomic analysis differentiates NEC versus no NEC in the domains of tissue injury, glucose uptake, and chemokine signaling.


Subject(s)
Enterocolitis, Necrotizing/veterinary , Fat Emulsions, Intravenous/pharmacology , Fatty Acids/metabolism , Ileum/drug effects , Metabolome , Animals , Enterocolitis, Necrotizing/chemically induced , Humans , Ileum/metabolism , Parenteral Nutrition/adverse effects , Premature Birth , Risk Factors , Swine , Swine Diseases/chemically induced
10.
J Nutr ; 149(10): 1724-1731, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31179494

ABSTRACT

BACKGROUND: Developmental expression of fatty acid transporters and their role in polyunsaturated fatty acid concentrations in the postnatal period have not been evaluated. OBJECTIVE: We hypothesized that transporter expression is developmentally regulated, tissue-specific, and that expression can modulate fatty acid accretion independently of diet. METHODS: Brain and lung transporter expression were quantified in C57BL/6 wild-type (WT) and Fat1 mice. Pups were dam-fed until day 21. Dams were fed AIN-76A 10% corn oil to represent a typical North American/European diet. After weaning, mice were fed the same diet as dams. Gene expression of Fatp1, Fatp4, Fabp5, and Fat/cd36 was quantified by quantitative reverse transcriptase-polymerase chain reaction. Fatty acid concentrations were measured by GC-MS. RESULTS: Brain docosahexaenoic acid (DHA) concentrations increased from day 3 to day 28 in both genotypes, with higher concentrations at days 3 and 14 in Fat1 than in WT mice [median (IQR)]: 10.7 (10.6-11.2) mol% compared with 6.6 (6.4-7.2) mol% and 12.5 (12.4-12.9) mol% compared with 8.9 (8.7-9.1) mol%, respectively; P < 0.05). During DHA accrual, transporter expression decreased. Fold changes in brain Fatp4, Fabp5, and Fat/cd36 were inversely correlated with fold changes in brain DHA concentrations in Fat1 relative to WT mice (ρ = -0.85, -0.75, and -0.78, respectively; P ≤ 0.001). Lung DHA concentrations were unchanged across the 3 time points for both genotypes. Despite unchanging DHA concentrations, there was increased expression of Fatp1 at days 14 and 28 (5-fold), Fatp4 at day 14 (2.3-fold), and Fabp5 at day 14 (3.8-fold) relative to day 3 in Fat1 mice. In WT mice, Fatp1 increased almost 5-fold at day 28 relative to day 3. There was no correlation between lung transporters and DHA concentrations in Fat1 relative to WT mice. CONCLUSIONS: Development of fatty acid transporter expression in C57BL/6 WT and Fat1 mice is genotype and tissue specific. Further, postnatal accretion of brain DHA appears independent of transporter status, with tissue concentrations representing dietary contributions.


Subject(s)
Brain/metabolism , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/pharmacology , Fatty Acid Transport Proteins/metabolism , Lung/metabolism , Animals , Corn Oil/administration & dosage , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Docosahexaenoic Acids/metabolism , Fatty Acid Transport Proteins/genetics , Fatty Acids/metabolism , Female , Gene Expression Regulation/drug effects , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/isolation & purification
12.
Pediatr Res ; 85(4): 556-565, 2019 03.
Article in English | MEDLINE | ID: mdl-30653193

ABSTRACT

BACKGROUND: Long-chain polyunsaturated fatty acids (LCPUFAs) play a critical role in neonatal health. We hypothesized that LCPUFAs play an essential role in priming postnatal gut development. We studied the effect of LCPUFAs on postnatal gut development using fat-1 transgenic mice, which are capable of converting n-6 to n-3 LCPUFAs, and wild-type (WT) C57BL/6 mice. METHODS: Distal ileum sections were collected from fat-1 and WT mice on days 3, 14, and 28. Fatty acid analyses, histology, RT-qPCR and intestinal permeability were performed. RESULTS: Fat-1 mice, relative to WT mice, showed increased n-3 LCPUFAs levels (α-linolenic acid, docosahexaenoic acid, and eicosapentaenoic acid, p < 0.05) and decreased arachidonic acid levels (p < 0.05) in the ileum. Preweaning fat-1 mice, compared to WT, showed >50% reduced muc2, Tff3, TLR9, and Camp expression (p < 0.05), markers of the innate immune response. There was a >two-fold increased expression of Fzd5 and EphB2, markers of cell differentiation (p < 0.05), and Fabp2 and 6, regulators of fatty acid transport and metabolism (p < 0.05). Despite reduced expression of tight junction genes, intestinal permeability in fat-1 was comparable to WT mice. CONCLUSIONS: Our data support the hypothesis that fatty acid profiles early in development modulate intestinal gene expression in formative domains, such as cell differentiation, tight junctions, other innate host defenses, and lipid metabolism.


Subject(s)
Cadherins/genetics , Fatty Acids, Unsaturated/pharmacology , Ileum/drug effects , Animals , Fatty Acids/metabolism , Gene Expression Regulation, Developmental/drug effects , Ileum/growth & development , Ileum/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects
14.
J Pediatr Gastroenterol Nutr ; 67(1): 97-102, 2018 07.
Article in English | MEDLINE | ID: mdl-29620597

ABSTRACT

OBJECTIVES: The primary objective of this study was to evaluate early postnatal serum gut hormone concentrations in preterm infants as predictors of time to full enteral feedings. The secondary objective was to identify infant characteristics and nutritional factors that modulate serum gut hormone concentrations and time to full enteral feedings. METHODS: Sixty-four preterm infants less than 30 weeks of gestation were included in this retrospective cohort study. Serum gut hormone concentrations at postnatal days 0 and 7 were measured using enzyme-linked immunosorbent assays. Linear regression and mediation analyses were performed. RESULTS: Median (interquartile range) serum concentrations of glucose-dependent insulinotropic peptide (GIP) and peptide YY (PYY) on postnatal day 7 were 31.3 pg/mL (18.2, 52.3) and 1181.7 pg/mL (859.0, 1650.2), respectively. GIP and PYY concentrations on day 7 were associated with days to full enteral feedings after adjustment for confounders (ß = -1.1, P = 0.03; and ß = -0.002, P = 0.02, respectively). Nutritional intake was correlated with serum concentrations of GIP and PYY on postnatal day 7 and time to full enteral feedings. Mediation analysis revealed that the effect of serum gut hormone concentrations on time to full enteral feedings was not fully explained by nutritional intake. Intrauterine growth restriction, mechanical ventilation on postnatal day 7, and patent ductus arteriosus treated with indomethacin were associated with longer time to full enteral feedings. CONCLUSIONS: Serum concentrations of GIP and PYY on postnatal 7 are independently associated with time to full enteral feedings. The link between serum gut hormone concentrations and time to full enteral feedings is not fully mediated by nutritional factors, suggesting an independent mechanism underlying the influence of gut hormones on feeding tolerance and time to full enteral feedings.


Subject(s)
Enteral Nutrition , Gastric Inhibitory Polypeptide/blood , Gastrointestinal Tract/growth & development , Infant, Premature/growth & development , Peptide YY/blood , Cardiovascular Agents/therapeutic use , Ductus Arteriosus, Patent/drug therapy , Female , Fetal Growth Retardation/physiopathology , Humans , Indomethacin/therapeutic use , Infant, Newborn , Male , Nutritional Physiological Phenomena , Respiration, Artificial , Retrospective Studies , Time Factors
15.
J Perinat Med ; 47(1): 106-113, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-29995634

ABSTRACT

Background Mothers of preterm (PT) infants have difficulty providing adequate quantities of human milk (HM) for their babies during their hospital stay. The macronutrient content in HM changes over time, varying across and within individual mothers. The research aim of the strudy was to describe the intake of mothers' own milk (MOM) and its composition according to gestational (GA) and postnatal age (PNA) in infants born <32 weeks' GA and to correlate them with neonatal weight, length and morbidities. Methods A prospective observational study of 176 premature infants in a unit without a donor milk bank was conducted. Daily milk intake was recorded. HM macronutrients were determined by mid-infrared spectrophotometric analysis at 7, 15 and 30 days after delivery and monthly until hospital discharge. Results Intake of MOM increased during the first 2 weeks after birth and decreased steadily thereafter. Protein concentration varied inversely with PNA. Carbohydrate and lipid concentrations increased over the first few days and remained stable thereafter. A fall in weight percentiles from birth to 60 days was found. No correlation was found between total protein and calorie intakes at 3 and 15 days of life and growth velocity (GV) between 15 and 30 days, even when broken down into parenteral nutrition (PN), formula and MOM. Conclusion To improve MOM feeding in PT newborns, intensive support strategies at the prenatal stage along entire hospitalization income should be encouraged. New protocols for fortification of HM should be implemented to optimize postnatal weight gain while preserving the health benefits of HM.


Subject(s)
Infant, Premature , Milk Banks , Milk, Human , Nutrients , Weight Gain/physiology , Body Weight , Breast Feeding , Female , Gestational Age , Hospitalization , Humans , Infant Nutritional Physiological Phenomena , Infant, Newborn , Infant, Premature/growth & development , Infant, Premature/physiology , Male , Nutritional Requirements , Prospective Studies , Spain , Spectrum Analysis/methods
16.
BMC Med ; 15(1): 29, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183317

ABSTRACT

BACKGROUND: Exocrine pancreatic insufficiency (EPI) is characterized by a deficiency of exocrine pancreatic enzymes, resulting in malabsorption. Numerous conditions account for the etiology of EPI, with the most common being diseases of the pancreatic parenchyma including chronic pancreatitis, cystic fibrosis, and a history of extensive necrotizing acute pancreatitis. Treatment for EPI includes dietary management, lifestyle changes (i.e., decrease in alcohol consumption and smoking cessation), and pancreatic enzyme replacement therapy. DISCUSSION: Many diagnostic tests are available to diagnose EPI, however, the criteria of choice remain unclear and the causes for a false-positive test are not yet understood. Despite multiple studies on the treatment of EPI using exogenous pancreatic enzymes, there remains confusion amongst medical practitioners with regard to the best approach to diagnose EPI, as well as dosing and administration of pancreatic enzymes. Appropriate use of diagnostics and treatment approaches using pancreatic enzymes in EPI is essential for patients. This opinion piece aims to address the existing myths, remove the current confusion, and function as a practical guide to the diagnosis and treatment of EPI.


Subject(s)
Exocrine Pancreatic Insufficiency/therapy , Pancreas/physiopathology , Humans
17.
Pancreatology ; 17(2): 176-181, 2017.
Article in English | MEDLINE | ID: mdl-28237616

ABSTRACT

BACKGROUND/OBJECTIVES: Hyperlipasemia is frequently encountered in patients in the intensive care unit (ICU). The degree to which it should be valued in making the diagnosis of acute pancreatitis (AP) in critically ill patients remains uncertain. We sought to determine the diagnostic accuracy of hyperlipasemia and the optimal lipase cutoff for diagnosing AP in critically ill patients. METHODS: Four hundred and seventeen ICU patients with hyperlipasemia, defined as lipase greater than three times the upper limit of normal from 2009 to 2012 were retrospectively identified. A diagnosis of AP was confirmed by the additional presence of either characteristic abdominal pain or cross-sectional imaging. RESULTS: The overall positive predictive value (PPV) of hyperlipasemia was 38.1%. Median initial lipase levels were 1164 IU/L in patients with AP and 284.5 IU/L in patients without AP (p < 0.001). The optimal diagnostic lipase cutoff of 532 IU/L correlated with a sensitivity, specificity, negative predictive value and PPV of 77.4%, 78.0%, 84.9%, and 67.0% respectively. The most common primary diagnoses in non-AP patients with elevated lipase included shock, cardiac arrest and malignancy. CONCLUSIONS: Physicians should maintain caution when interpreting hyperlipasemia in the critically ill due its relatively low PPV. However, a greater lipase cutoff improves its diagnostic value in AP and helps to reduce unnecessary imaging in these patients.


Subject(s)
Critical Illness , Lipase/blood , Pancreatitis/diagnosis , Aged , Female , Humans , Male , Middle Aged , Pancreatitis/blood , Predictive Value of Tests , Sensitivity and Specificity
19.
Pediatr Res ; 82(5): 829-838, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28665922

ABSTRACT

BackgroundThe preterm infant gut microbiota is vulnerable to different biotic and abiotic factors. Although the development of this microbiota has been extensively studied, the mobilome-i.e. the mobile genetic elements (MGEs) in the gut microbiota-has not been considered. Therefore, the aim of this study was to investigate the association of the mobilome with birth weight and hospital location in the preterm infant gut microbiota.MethodsThe data set consists of fecal samples from 62 preterm infants with and without necrotizing enterocolitis (NEC) from three different hospitals. We analyzed the gut microbiome by using 16S rRNA amplicon sequencing, shot-gun metagenome sequencing, and quantitative PCR. Predictive models and other data analyses were performed using MATLAB and QIIME.ResultSThe microbiota composition was significantly different between NEC-positive and NEC-negative infants and significantly different between hospitals. An operational taxanomic unit (OTU) showed strong positive and negative correlation with NEC and birth weight, respectively, whereas none showed significance for mode of delivery. Metagenome analyses revealed high levels of conjugative plasmids with MGEs and virulence genes. Results from quantitative PCR showed that the plasmid signature genes were significantly different between hospitals and in NEC-positive infants.ConclusionOur results point toward an association of the mobilome with hospital location in preterm infants.


Subject(s)
Birth Weight , DNA, Bacterial/genetics , Enterocolitis, Necrotizing/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Infant, Premature , Infant, Very Low Birth Weight , Interspersed Repetitive Sequences , Premature Birth/microbiology , Case-Control Studies , Computational Biology , Databases, Genetic , Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/epidemiology , Feces/microbiology , Female , Genome, Bacterial , Gestational Age , Humans , Infant, Newborn , Male , Metagenome , Metagenomics/methods , Premature Birth/diagnosis , Premature Birth/epidemiology , Ribotyping , United States/epidemiology
20.
J Lipid Res ; 57(9): 1696-711, 2016 09.
Article in English | MEDLINE | ID: mdl-27474222

ABSTRACT

We aimed to characterize the lipidomic, metabolomic, and transcriptomic profiles in preterm piglets administered enteral (ENT) formula or three parenteral lipid emulsions [parenteral nutrition (PN)], Intralipid (IL), Omegaven (OV), or SMOFlipid (SL), for 14 days. Piglets in all parenteral lipid groups showed differential organ growth versus ENT piglets; whole body growth rate was lowest in IL piglets, yet there were no differences in either energy expenditure or (13)C-palmitate oxidation. Plasma homeostatic model assessment of insulin resistance demonstrated insulin resistance in IL, but not OV or SL, compared with ENT. The fatty acid and acyl-CoA content of the liver, muscle, brain, and plasma fatty acids reflected the composition of the dietary lipids administered. Free carnitine and acylcarnitine (ACT) levels were markedly reduced in the PN groups compared with ENT piglets. Genes associated with oxidative stress and inflammation were increased, whereas those associated with alternative pathways of fatty acid oxidation were decreased in all PN groups. Our results show that new generation lipid emulsions directly enrich tissue fatty acids, especially in the brain, and lead to improved growth and insulin sensitivity compared with a soybean lipid emulsion. In all total PN groups, carnitine levels are limiting to the formation of ACTs and gene expression reflects the stress of excess lipid on liver function.


Subject(s)
Fish Oils/administration & dosage , Lipid Metabolism/genetics , Phospholipids/administration & dosage , Soybean Oil/administration & dosage , Triglycerides/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Dietary Fats, Unsaturated/administration & dosage , Emulsions/administration & dosage , Fatty Acids/metabolism , Liver/drug effects , Liver/metabolism , Male , Palmitic Acid/administration & dosage , Palmitic Acid/metabolism , Parenteral Nutrition , Swine/growth & development , Swine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL