Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Pediatr Emerg Care ; 38(3): e1087-e1089, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34507345

ABSTRACT

ABSTRACT: Amyand hernia is a rare type of inguinal hernia defined by the presence of the appendix in the inguinal hernia sac. Clinical diagnosis of Amyand hernia can be challenging because this diagnosis is typically made intraoperatively, often as an incidental finding. Preoperative diagnosis by computed tomography and radiology ultrasound has previously been reported; however, there exists no reports of the diagnosis being made by point-of-care ultrasound. We present a case of Amyand hernia visible on point-of-care ultrasound performed by a pediatric emergency medicine physician.


Subject(s)
Appendix , Hernia, Inguinal , Appendix/diagnostic imaging , Child , Hernia, Inguinal/diagnostic imaging , Hernia, Inguinal/surgery , Humans , Point-of-Care Systems , Point-of-Care Testing , Ultrasonography
2.
J Pediatr ; 227: 69-76.e3, 2020 12.
Article in English | MEDLINE | ID: mdl-32687916

ABSTRACT

OBJECTIVES: To assess whether a 24-hour length of hospitalization and empiric antibiotic therapy to exclude central line-associated bloodstream infection (CLABSI) in children with intestinal failure is potentially as safe as 48 hours, which is the duration most commonly used but not evidence based. STUDY DESIGN: A prospective single-institution observational cohort study was conducted among pediatric patients with intestinal failure from July 1, 2015, through June 30, 2018, to identify episodes of suspected CLABSI. The primary end point was time from blood sampling to positive blood culture. Secondary end points included presenting symptoms, laboratory test results, responses to a parent/legal guardian-completed symptom survey, length of inpatient stay, costs, and charges. RESULTS: Seventy-three patients with intestinal failure receiving nutritional support via central venous catheters enrolled; 35 were hospitalized with suspected CLABSI at least once during the study. There were 49 positive blood cultures confirming CLABSI in 128 episodes (38%). The median time from blood sampling to positive culture was 11.1 hours. The probability of a blood culture becoming positive after 24 hours was 2.3%. Elevated C-reactive protein and neutrophil predominance in white blood cell count were associated with positive blood cultures. Estimated cost savings by transitioning from a 48-hour to a 24-hour admission to rule-out CLABSI was $4639 per admission. CONCLUSIONS: A 24-hour duration of empiric management to exclude CLABSI may be appropriate for patients with negative blood cultures and no clinically concerning signs. A multi-institutional study would more robustly differentiate patients safe for discharge after 24 hours from those who warrant longer empiric treatment.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Catheter-Related Infections/prevention & control , Catheterization, Central Venous/adverse effects , Intestinal Diseases/therapy , Anti-Bacterial Agents/adverse effects , C-Reactive Protein/analysis , Case-Control Studies , Catheter-Related Infections/blood , Catheter-Related Infections/diagnosis , Catheter-Related Infections/economics , Catheterization, Central Venous/instrumentation , Catheters, Indwelling/microbiology , Child , Child, Preschool , Female , Humans , Infant , Intestinal Diseases/economics , Length of Stay/economics , Length of Stay/statistics & numerical data , Male , Parenteral Nutrition/adverse effects , Parenteral Nutrition/methods , Prospective Studies , Surveys and Questionnaires , Time Factors
3.
JPEN J Parenter Enteral Nutr ; 44(5): 951-958, 2020 07.
Article in English | MEDLINE | ID: mdl-31282035

ABSTRACT

BACKGROUND: Vitamin K is a fat-soluble compound that plays important roles in coagulation. In children with intestinal failure-associated liver disease (IFALD), the disrupted enterohepatic circulation can lead to intestinal loss of vitamin K. Fish oil-based lipid emulsion (FOLE) has proven effective in treating IFALD. As biliary excretion is restored during cholestasis reversal, the accelerated vitamin K loss can pose a risk for deficiency. METHODS: Ten neonates with IFALD and receiving FOLE monotherapy were prospectively enrolled in the study from 2016 to 2018. In addition to weekly measurements of international normalized ratio (INR) and direct bilirubin (DB), ostomy output was collected for determination of fecal concentrations of phylloquinone (PK). Trends of DB, INR, and fecal PK concentrations were summarized with locally estimated scatterplot smoothing. RESULTS: The median time (interquartile range) from FOLE initiation to cholestasis reversal was 59 (19-78) days. During cholestasis reversal, INR remained relatively unchanged, whereas the mean (95% confidence interval) daily fecal excretion of PK increased from 25.1 (5.0-158.5) ng at the time of FOLE initiation to 158.5 (31.6-1000.0) ng at complete reversal. Examination of individual trends in fecal PK excretion and INR revealed little correlation between the 2 measurements (r = -0.10; P = 0.50). CONCLUSION: Children with IFALD are at risk for vitamin K deficiency during cholestasis reversal. Close monitoring and quantified supplementation of vitamin K may be warranted during this period. However, this should not be guided by INR alone, as it is a poor indicator of vitamin K status.


Subject(s)
Cholestasis , Vitamin K , Child , Cholestasis/drug therapy , Cholestasis/etiology , Fat Emulsions, Intravenous , Humans , Infant, Newborn , International Normalized Ratio , Male , Parenteral Nutrition
4.
PLoS One ; 14(7): e0217155, 2019.
Article in English | MEDLINE | ID: mdl-31295333

ABSTRACT

Intestinal failure-associated liver disease (IFALD) is a risk of parenteral nutrition (PN)-dependence. Intravenous soybean oil-based parenteral fat can exacerbate the risk of IFALD while intravenous fish oil can minimize its progression, yet the mechanisms by which soybean oil harms and fish oil protects the liver are uncertain. Properties that differentiate soybean and fish oils include α-tocopherol and phytosterol content. Soybean oil is rich in phytosterols and contains little α-tocopherol. Fish oil contains abundant α-tocopherol and little phytosterols. This study tested whether α-tocopherol confers hepatoprotective properties while phytosterols confer hepatotoxicity to intravenous fat emulsions. Utilizing emulsions formulated in the laboratory, a soybean oil emulsion (SO) failed to protect from hepatosteatosis in mice administered a PN solution enterally. An emulsion of soybean oil containing α-tocopherol (SO+AT) preserved normal hepatic architecture. A fish oil emulsion (FO) and an emulsion of fish oil containing phytosterols (FO+P) protected from steatosis in this model. Expression of hepatic acetyl CoA carboxylase (ACC) and peroxisome proliferator-activated receptor gamma (PPARγ), was increased in animals administered SO. ACC and PPARγ levels were comparable to chow-fed controls in animals receiving SO+AT, FO, and FO+P. This study suggests a hepatoprotective role for α-tocopherol in liver injury induced by the enteral administration of a parenteral nutrition solution. Phytosterols do not appear to compromise the hepatoprotective effects of fish oil.


Subject(s)
Fat Emulsions, Intravenous/therapeutic use , Fatty Liver/etiology , Fatty Liver/prevention & control , Protective Agents/therapeutic use , alpha-Tocopherol/therapeutic use , Animals , Disease Models, Animal , Fat Emulsions, Intravenous/administration & dosage , Fat Emulsions, Intravenous/adverse effects , Fatty Liver/pathology , Fish Oils/administration & dosage , Fish Oils/adverse effects , Fish Oils/therapeutic use , Mice, Inbred C57BL , Parenteral Nutrition/adverse effects , Phytosterols/administration & dosage , Phytosterols/adverse effects , Phytosterols/therapeutic use , Protective Agents/administration & dosage , Protective Agents/adverse effects , Soybean Oil/administration & dosage , Soybean Oil/adverse effects , Soybean Oil/therapeutic use , alpha-Tocopherol/administration & dosage , alpha-Tocopherol/adverse effects
5.
J Pediatr Surg ; 54(11): 2392-2397, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31036368

ABSTRACT

BACKGROUND: A single dose of IV fish oil (FO) before hepatic ischemia reperfusion injury (HIRI) increases hepatocyte proliferation and reduces necrosis in wild type (WT) mice. It has been suggested that the GPR120 receptor on Kupffer cells mediates FO's ability to reduce HIRI. The purpose of this study was to determine whether GPR120 is required for FO to reduce HIRI. METHODS: Sixty-four (n = 8/group) adult male WT (C57BL/6) and GPR120 knockout (KO) mice received IV FO (1 g/kg) or saline 1 h prior to HIRI or sham operation. Mice were euthanized 24 h postoperatively for analysis of hepatic histology, NFκB activity, and serum alanine transaminase (ALT) levels. RESULTS: FO pretreated livers had less necrosis after HIRI than saline pretreated livers in both WT (mean ±â€¯SEM 25.9 ±â€¯7.3% less, P = 0.007) and KO (36.6 ±â€¯7.3% less, P < 0.0001) mice. There was no significant difference in percent necrosis between WT-FO and KO-FO groups. Sham groups demonstrated minimal necrosis (0-1.9%). Mean [95% CI] ALT after HIRI was significantly higher (P = 0.04) in WT-Saline mice (1604 U/L [751-3427]) compared to WT-FO (321 U/L [150-686]) but was not significantly higher in KO-Saline mice compared to KO-FO. There were no differences in ALT between WT-FO and KO-FO mice who underwent HIRI or between groups who underwent sham surgery. There were no differences in NFκB or IKKß activation among groups as measured by Western blot analysis. CONCLUSIONS: IV FO pretreatment was able to reduce HIRI in GPR120 KO mice, suggesting the hepatoprotective effects of FO are not mediated by GPR120 alone.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Receptors, G-Protein-Coupled/drug effects , Reperfusion Injury/prevention & control , Alanine Transaminase/blood , Animals , Cell Proliferation , Hepatocytes/cytology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p50 Subunit/metabolism , Necrosis/pathology , Reperfusion Injury/pathology , Signal Transduction/drug effects
6.
Article in English | MEDLINE | ID: mdl-30975380

ABSTRACT

Intravenous fish oil lipid emulsions (FOLE) can prevent parenteral nutrition (PN)-induced liver injury in murine models and reverse PN-induced cholestasis in pediatric patients. However, the mechanisms by which fish oil protects the liver are incompletely characterized. Fish oil is rich in omega-3 fatty acids, which are ligands for the G-protein coupled receptor 120 (GPR120), expressed on hepatic Kupffer cells. This study tested the hypothesis that FOLE protects the liver from PN-induced injury through GPR120 signaling. Utilizing a previously described murine model of PN-induced liver injury in which mice develop steatosis in response to an oral parenteral nutrition diet, FOLE was able to preserve normal hepatic architecture in wild type mice, but not in congenic GPR120 knockout (gpr120-/-) mice. To further characterize the requirement of intact GPR120 for FOLE-mediated hepatic protection, gene expression profiles of key regulators of fat metabolism were measured. PPARγ was identified as a gene that is up-regulated by the PN diet and normalized with the addition of FOLE in wild type, but not in gpr120-/- mice. This was confirmed at the protein expression level. A PPARγ expression array further identified CD36 and SCD1, both down-stream effectors of PPARγ, to be up-regulated in PN-fed wild type mice yet normalized upon FOLE administration in wild type but not in gpr120-/- mice. Together, these results suggest that FOLE protects the liver, in part, through activation of GPR120 and the downstream effectors PPARγ and CD36. Identification of key genetic determinants of FOLE-mediated hepatic protection may provide targets for small molecule-based hepatic protection strategies.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Fat Emulsions, Intravenous/therapeutic use , Fish Oils/therapeutic use , PPAR gamma/metabolism , Parenteral Nutrition/adverse effects , Protective Agents/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Animals , CD36 Antigens/metabolism , Disease Models, Animal , Fat Emulsions, Intravenous/administration & dosage , Fatty Liver/drug therapy , Fish Oils/administration & dosage , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Stearoyl-CoA Desaturase/metabolism
7.
Front Neurosci ; 13: 72, 2019.
Article in English | MEDLINE | ID: mdl-30858795

ABSTRACT

Polyunsaturated fatty acids serve multiple functions in neurodevelopment and neurocognitive function. Intravenous lipid emulsions are administered to children that are dependent on parenteral nutrition to provide the essential fatty acids needed to sustain growth and development. One of these emulsions, derived from fish-oil, is particularly poor in the traditional essential fatty acids, linoleic and alpha-linolenic acids. However, it does contain adequate amounts of its main derivatives, arachidonic acid (ARA) and docosahexaenoic acid (DHA), respectively. This skewed composition has raised concern about the sole use of fish-oil based lipid emulsions in children and how its administration can be detrimental to their neurodevelopment. Using a custom-made diet that contains ARA and DHA as a sole source of polyunsaturated fatty acids, we bred and fed mice for multiple generations. Compared to adult, chow-fed mice, animals maintained on this special diet showed similar outcomes in a battery of neurocognitive tests performed under controlled conditions. Chow-fed mice did perform better in the rotarod test for ataxia and balance, although both experimental groups showed a conserved motor learning capacity. Conversely, mice fed the custom diet rich in DHA and ARA showed less neophobia than the chow-fed animals. Results from these experiments suggest that providing a diet where ARA and DHA are the sole source of polyunsaturated fatty acids is sufficient to support gross visual, cognitive, motor, and social development in mice.

8.
Am J Clin Nutr ; 109(4): 1038-1050, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30882140

ABSTRACT

BACKGROUND: Fish oil (FO) intravenous lipid emulsions (ILEs) are used as a monotherapy to treat parenteral nutrition (PN)-associated liver disease and provide essential fatty acids (EFAs) needed to sustain growth and prevent EFA deficiency (EFAD). Studies have suggested that medium-chain triglycerides (MCTs) and α-tocopherol have anti-inflammatory properties. OBJECTIVE: The purpose of this study was to test whether FO-ILEs containing MCTs and/or additional α-tocopherol decrease the inflammatory response to an endotoxin challenge compared with FO-ILE alone and preserve the ability to prevent PN-induced liver injury in mice. METHODS: A murine model of PN-induced hepatosteatosis was used to compare the effects of ILEs formulated in the laboratory containing varying ratios of FO and MCTs, and subsequently FO- and 50:50 FO:MCT-ILE plus 500 mg/L α-tocopherol (FO + AT and 50:50 + AT, respectively). C57BL/6 mice receiving unpurified diet (UPD), PN-equivalent diet (PN) + saline, and PN + soybean oil (SO)-ILE served as controls. After 19 d, mice received an intraperitoneal saline or endotoxin challenge 4 h before being killed. Serum and livers were harvested for histologic analysis, fatty acid profiling, and measurement of systemic inflammatory markers (tumor necrosis factor-α, interleukin-6). RESULTS: All ILEs were well tolerated and prevented biochemical EFAD. Livers of mice that received saline and SO developed steatosis. Mice that received 30:70 FO:MCT developed mild hepatosteatosis. All other FO-containing ILEs preserved normal hepatic architecture. Mice that received FO- or SO-ILE had significantly elevated systemic inflammatory markers after endotoxin challenge compared with UPD-fed controls, whereas 50:50 FO:MCT, 30:70 FO:MCT, FO + AT, and 50:50 + AT groups had significantly lower inflammatory markers similar to those seen in UPD-fed controls. CONCLUSIONS: Mixed FO/MCT and the addition of α-tocopherol to FO improved the inflammatory response to endotoxin challenge compared with FO-ILE alone while still preventing PN-induced liver injury and EFAD in mice. There was no synergistic relation between α-tocopherol and MCTs.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Fat Emulsions, Intravenous/administration & dosage , Fish Oils/administration & dosage , Liver Diseases/prevention & control , Parenteral Nutrition/adverse effects , Triglycerides/administration & dosage , Triglycerides/chemistry , alpha-Tocopherol/administration & dosage , Animals , Anti-Inflammatory Agents/chemistry , Disease Models, Animal , Fat Emulsions, Intravenous/chemistry , Fish Oils/chemistry , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Liver Diseases/etiology , Liver Diseases/genetics , Liver Diseases/immunology , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Clin Nutr ; 38(5): 2145-2154, 2019 10.
Article in English | MEDLINE | ID: mdl-30224305

ABSTRACT

BACKGROUND & AIMS: Altering the lipid component in diets may affect the incidence of metabolic bone disease in patients dependent on parenteral nutrition. Consumption of polyunsaturated fatty acids (PUFA) can impact bone health by modulating calcium metabolism, prostaglandin synthesis, lipid oxidation, osteoblast formation, and osteoclastogenesis. The aim of this study was to evaluate the dietary effects of PUFA on murine bone health. METHODS: Three-weeks-old male (n = 30) and female (n = 30) C57BL/6J mice were randomized into one of three dietary groups. The diets differed only in fat composition: soybean oil (SOY), rich in ω-6 PUFA; docosahexaenoic acid alone (DHA), an ω-3 PUFA; and DHA with arachidonic acid, an ω-6 PUFA, at a 20:1 ratio (DHA/ARA). After 9 weeks of dietary treatment, femurs were harvested for micro-computed tomographic analysis and mechanical testing via 3-point bending. Separate mice from each group were used solely for serial blood draws for measurement of biomarkers of bone formation and resorption. RESULTS: At the microstructural level, although some parameters in cortical bone reached differences that were statistically significant in female mice, these were too small to be considered biologically relevant. Similarly, trabecular bone parameters in male mice were statistically different in some dietary groups, although the biological interpretation of such subtle changes translate into a lack of effect in favor of any of the experimental diets. No differences were noted at the mechanical level and in blood-based biomarkers of bone metabolism across dietary groups within gender. CONCLUSIONS: Subtle differences were noted at the bones' microstructural level, however these are likely the result of random effects that do not translate into changes that are biologically relevant. Similarly, differences were not seen at the mechanical level, nor were they reflected in blood-based biomarkers of bone metabolism. Altogether, dietary consumption of PUFA do not seem to affect bone structure or metabolism in a healthy model of growing mice.


Subject(s)
Cancellous Bone , Fatty Acids, Omega-3 , Femur , Animals , Bone Density/drug effects , Cancellous Bone/chemistry , Cancellous Bone/cytology , Cancellous Bone/drug effects , Cancellous Bone/physiology , Diet , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/pharmacology , Female , Femur/chemistry , Femur/drug effects , Femur/physiology , Male , Mice , Mice, Inbred C57BL , Weight Gain
10.
PLoS One ; 13(12): e0208579, 2018.
Article in English | MEDLINE | ID: mdl-30566445

ABSTRACT

Exogenous vascular endothelial growth factor (VEGF) accelerates compensatory lung growth (CLG) in mice after unilateral pneumonectomy. In this study, we unexpectedly discovered a method to enhance CLG with a VEGF inhibitor, soluble VEGFR1. Eight-week-old C57BL/6 male mice underwent left pneumonectomy, followed by daily intraperitoneal (ip) injection of either saline (control) or 20 µg/kg of VEGFR1-Fc. On post-operative day (POD) 4, mice underwent pulmonary function tests (PFT) and lungs were harvested for volume measurement and analyses of the VEGF signaling pathway. To investigate the role of hypoxia in mediating the effects of VEGFR1, experiments were repeated with concurrent administration of PT-2385, an inhibitor of hypoxia-induced factor (HIF)2α, via orogastric gavage at 10 mg/kg every 12 hours for 4 days. We found that VEGFR1-treated mice had increased total lung capacity (P = 0.006), pulmonary compliance (P = 0.03), and post-euthanasia lung volume (P = 0.049) compared to control mice. VEGFR1 treatment increased pulmonary levels of VEGF (P = 0.008) and VEGFR2 (P = 0.01). It also stimulated endothelial proliferation (P < 0.0001) and enhanced pulmonary surfactant production (P = 0.03). The addition of PT-2385 abolished the increase in lung volume and endothelial proliferation in response to VEGFR1. By paradoxically stimulating angiogenesis and enhancing lung growth, VEGFR1 could represent a new treatment strategy for neonatal lung diseases characterized by dysfunction of the HIF-VEGF pathway.


Subject(s)
Lung/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Dose-Response Relationship, Drug , Half-Life , Lung/growth & development , Lung/physiology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Pneumonectomy , Recombinant Fusion Proteins/biosynthesis , Respiratory Function Tests , Signal Transduction/drug effects , Surface-Active Agents/metabolism , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Surgery ; 164(6): 1279-1286, 2018 12.
Article in English | MEDLINE | ID: mdl-30193736

ABSTRACT

BACKGROUND: Vascular endothelial growth factor has been found to accelerate compensatory lung growth after left pneumonectomy in mice. The aim of this study was to determine the natural history and the effects of vascular endothelial growth factor on compensatory lung growth in a large animal model. METHODS: To determine the natural history of compensatory lung growth, female Yorkshire piglets underwent a left pneumonectomy on days of life 10-11. Tissue harvest and volume measurement of the right lung were performed at baseline (n = 5) and on postoperative days 7 (n = 5), 14 (n = 4), and 21 (n = 5). For pharmacokinetic studies, vascular endothelial growth factor was infused via a central venous catheter, with plasma vascular endothelial growth factor levels measured at various time points. To test the effect of vascular endothelial growth factor on compensatory lung growth, 26 female Yorkshire piglets underwent a left pneumonectomy followed by daily infusion of vascular endothelial growth factor at 200 µg/kg or isovolumetric 0.9% NaCl (saline control). Lungs were harvested on postoperative day 7 for volume measurement and morphometric analyses. RESULTS: Compared with baseline, right lung volume after left pneumonectomy increased by factors of 2.1 ± 0.6, 3.3 ± 0.6, and 3.6 ± 0.4 on postoperative days 7, 14, and 21, respectively. The half-life of VEGF ranged from 89 to 144 minutes. Lesser doses of vascular endothelial growth factor resulted in better tolerance, volume of distribution, and clearance. Compared with the control group, piglets treated with vascular endothelial growth factor had greater lung volume (P < 0.0001), alveolar volume (P = 0.001), septal surface area (P = 0.007) and total alveolar count (P = 0.01). CONCLUSION: Vascular endothelial growth factor enhanced alveolar growth in neonatal piglets after unilateral pneumonectomy.


Subject(s)
Lung/growth & development , Vascular Endothelial Growth Factor A/pharmacokinetics , Animals , Animals, Newborn , Biometry , Drug Evaluation, Preclinical , Female , Lung/drug effects , Pneumonectomy , Recombinant Proteins , Swine , Vascular Endothelial Growth Factor A/administration & dosage
12.
Angiogenesis ; 21(4): 837-848, 2018 11.
Article in English | MEDLINE | ID: mdl-29956017

ABSTRACT

Children with hypoplastic lung diseases, such as congenital diaphragmatic hernia, can require life support via extracorporeal membrane oxygenation and systemic anticoagulation, usually in the form of heparin. The role of heparin in angiogenesis and organ growth is inconclusive, with conflicting data reported in the literature. This study aimed to investigate the effects of heparin on lung growth in a model of compensatory lung growth (CLG). Compared to the absence of heparin, treatment with heparin decreased the vascular endothelial growth factor (VEGF)-mediated activation of VEGFR2 and mitogenic effect on human lung microvascular endothelial cells in vitro. Compared to non-heparinized controls, heparinized mice demonstrated impaired pulmonary mechanics, decreased respiratory volumes and flows, and reduced activity levels after left pneumonectomy. They also had lower lung volume, pulmonary septal surface area and alveolar density on morphometric analyses. Lungs of heparinized mice displayed decreased phosphorylation of VEGFR2 compared to the control group, with consequential downstream reduction in markers of cellular proliferation and survival. The use of bivalirudin, an alternative anticoagulant that does not interact with VEGF, preserved lung growth and pulmonary mechanics. These results demonstrated that heparin impairs CLG by reducing VEGFR2 activation. These findings raise concern for the clinical use of heparin in the setting of organ growth or regeneration.


Subject(s)
Heparin/pharmacology , Lung/growth & development , Pneumonectomy , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Hirudins/pharmacology , Humans , Lung/pathology , Male , Mice , Peptide Fragments/pharmacology , Recombinant Proteins/pharmacology
13.
PLoS One ; 13(6): e0198700, 2018.
Article in English | MEDLINE | ID: mdl-29879188

ABSTRACT

Vascular endothelial growth factor (VEGF) has previously been demonstrated to accelerate compensatory lung growth (CLG) in mice and may be a useful therapy for pulmonary hypoplasia. Systemic administration of VEGF can result in side effects such as hypotension and edema. The aim of this study was to explore nasal delivery as a route for intrapulmonary VEGF administration. Eight-week-old C57BL/6 male mice underwent left pneumonectomy, followed by daily nasal instillation of VEGF at 0.5 mg/kg or isovolumetric saline. Lung volume measurement, morphometric analysis, and protein expression studies were performed on lung tissues harvested on postoperative day (POD) 4. To understand the mechanism by which VEGF accelerates lung growth, proliferation of human bronchial epithelial cells (HBEC) was assessed in a co-culture model with lung microvascular endothelial cells (HMVEC-L) treated with and without VEGF (10 ng/mL). The assay was then repeated with a heparin-binding EGF-like growth factor (HB-EGF) neutralizing antibody ranging from 0.5-50 µg/mL. Compared to control mice, the VEGF-treated group displayed significantly higher lung volume (P = 0.001) and alveolar count (P = 0.005) on POD 4. VEGF treatment resulted in increased pulmonary expression of HB-EGF (P = 0.02). VEGF-treated HMVEC-L increased HBEC proliferation (P = 0.002) while the addition of an HB-EGF neutralizing antibody at 5 and 50 µg/mL abolished this effect (P = 0.01 and 0.002, respectively). These findings demonstrate that nasal delivery of VEGF enhanced CLG. These effects could be mediated by a paracrine mechanism through upregulation of HB-EGF, an epithelial cell mitogen.


Subject(s)
Endothelial Cells/metabolism , Epithelial Cells/metabolism , Lung/growth & development , Vascular Endothelial Growth Factor A/pharmacology , Administration, Intranasal , Animals , Endothelial Cells/cytology , Epithelial Cells/cytology , Humans , Lung/cytology , Male , Mice
14.
Pediatr Res ; 83(6): 1182-1189, 2018 06.
Article in English | MEDLINE | ID: mdl-29638228

ABSTRACT

BackgroundDeficiency of vascular endothelial growth factor (VEGF) is associated with hypoplastic lung diseases, such as congenital diaphragmatic hernia. Provision of VEGF has been demonstrated to be beneficial in hyperoxia-induced bronchopulmonary dysplasia, and hence could induce lung growth and improve the outcome in hypoplastic lung diseases. We aimed to determine the effects of exogenous VEGF in a rodent model of compensatory lung growth after left pneumonectomy.MethodsEight-to-ten-week-old C57Bl6 male mice underwent left pneumonectomy, followed by daily intra-peritoneal injections of saline or VEGF (0.5 mg/kg). Lung volume measurement, pulmonary function tests, and morphometric analyses were performed on post-operative day (POD) 4 and 10. The pulmonary expression of angiogenic factors was analyzed by quantitative polymerase chain reaction and western blot.ResultsLung volume on POD 4 was higher in the VEGF-treated mice (P=0.03). On morphometric analyses, VEGF increased the parenchymal volume (P=0.001), alveolar volume (P=0.0003), and alveolar number (P<0.0001) on POD 4. The VEGF group displayed higher levels of phosphorylated-VEGFR2/VEGFR2 (P=0.03) and epidermal growth factor (EGF) messenger RNA (P=0.01).ConclusionVEGF accelerated the compensatory lung growth in mice, by increasing the alveolar units. These changes may be mediated by VEGFR2 and EGF-dependent mechanisms.


Subject(s)
Lung/growth & development , Pulmonary Alveoli/growth & development , Vascular Endothelial Growth Factor A/metabolism , Animals , Bronchopulmonary Dysplasia/metabolism , Epidermal Growth Factor/metabolism , Hyperoxia/metabolism , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Organ Size , Organogenesis , Pneumonectomy , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
JPEN J Parenter Enteral Nutr ; 42(1): 71-77, 2018 01.
Article in English | MEDLINE | ID: mdl-29505154

ABSTRACT

BACKGROUND: The purpose of this study was to characterize fatty acid profiles (FAPs) in parenteral nutrition (PN)-dependent infants with intestinal failure-associated liver disease (IFALD) receiving soybean oil-based lipid emulsion (SO) doses of ∼3 and ∼1 g/kg/d. METHODS: Prospectively collected data were retrospectively reviewed. Serum FAPs of patients <1 year old who experienced development of IFALD while receiving standard PN with SO were examined before transitioning to a fish oil-based lipid emulsion for IFALD treatment. Time on SO, dose, gestational age, and weight- and length-for-age z scores were also reviewed. RESULTS: Among the 49 patients analyzed, there were no differences in demographics or anthropometrics between patients who received standard SO (SO-S) (n = 14, range of dosage 2.06-3.31 g/kg/d) and reduced SO (SO-R) (n = 35, range of dosage 0.90-1.34 g/kg/d). Patients received SO for a median of 53 days (interquartile range 39, 73) before FAP measurement. Patients who received SO-R had significantly higher Mead acid and lower α-linolenic, eicosapentaenoic, linoleic, stearic, total ω-3, and total ω-6 fatty acid levels than patients who received SO-S (P < .01). Triene:tetraene ratios were higher in patients who received SO-R (P = .0009), and no patients experienced biochemical essential fatty acid deficiency (EFAD). CONCLUSION: PN-dependent infants with IFALD receiving SO-R have different FAPs than patients receiving SO-S. No patients in either group had biochemical EFAD.


Subject(s)
Fat Emulsions, Intravenous/therapeutic use , Fatty Acids/blood , Fish Oils/therapeutic use , Intestinal Diseases/complications , Liver Diseases/etiology , Parenteral Nutrition , Soybean Oil/therapeutic use , Female , Humans , Infant , Intestinal Diseases/therapy , Male , Prospective Studies , Retrospective Studies
16.
Surgery ; 163(5): 1035-1039, 2018 05.
Article in English | MEDLINE | ID: mdl-29358007

ABSTRACT

BACKGROUND: Ischemia reperfusion injury is a barrier to liver surgery and transplantation, particularly for steatotic livers. The purpose of this study was to determine if pretreatment with a single dose of intravenous fish oil decreases hepatic ischemia reperfusion injury and improves recovery of injured livers. METHODS: Sixty adult male C57BL/6 mice received 1 g/kg intravenous fish oil (Omegaven, Fresenius Kabi) or isovolumetric 0.9% NaCl (saline) via tail vein 1 hour before 30 minutes of 70% hepatic ischemia. Animals were killed 4, 8, or 24 hours postreperfusion, and livers were harvested for histologic analysis. RESULTS: Four hours postreperfusion, saline-treated livers demonstrated marked ischemia diffusely around the central veins, while intravenous fish oil-treated livers demonstrated only patchy necrosis with intervening normal parenchyma. Eight hours postreperfusion, all livers demonstrated pale areas of cell loss with surrounding regenerating hepatocytes. Ki67 staining confirmed 14.4/10 high-powered field (95% confidence interval, 3.2-25.6) more regenerating hepatocytes around areas of necrosis in intravenous fish oil-treated livers. Twenty-four hours postreperfusion, all livers demonstrated patchy areas of necrosis, with an 89% (95% confidence interval, 85-92) decrease in the area of necrosis in intravenous fish oil-treated livers. CONCLUSION: Intravenous fish oil treatment prior to hepatic ischemia reperfusion injury decreased the area of hepatic necrosis and increased hepatocyte regeneration compared to saline treatment in a mouse model.


Subject(s)
Fish Oils/administration & dosage , Liver Diseases/prevention & control , Reperfusion Injury/prevention & control , Administration, Intravenous , Alanine Transaminase/blood , Animals , Body Weight , Disease Models, Animal , Drug Evaluation, Preclinical , Liver/pathology , Liver Diseases/blood , Liver Diseases/pathology , Mice, Inbred C57BL , Organ Size , Reperfusion Injury/blood , Reperfusion Injury/pathology
17.
Clin Nutr ; 37(3): 784-789, 2018 06.
Article in English | MEDLINE | ID: mdl-28716367

ABSTRACT

The essentiality of fatty acids was determined by the Burrs in the 1920s. It is commonly accepted that provision of linoleic (LA) and alpha-linolenic acids (ALA) prevents and reverses essential fatty acid deficiency (EFAD). Development of alternative injectable lipid emulsions (ILE) low in LA and ALA has raised concern about their ability to prevent EFAD. This review provides biochemical evidence coupled with observations from animal and human studies that aim to characterize which fatty acids are truly essential to prevent EFAD. Retroconversion pathways and mobilization from body stores suggest that arachidonic and docosahexaenoic acids (ARA and DHA - the main derivatives of LA and ALA, respectively) also prevent EFAD. Our group first proposed the essentiality of ARA and DHA by feeding mice exclusively these fatty acids and proving that they prevent EFAD. Survival for 5 generations on this diet provides additional evidence that growth and reproductive capabilities are maintained. Moreover, the use of fish oil-based ILE, with minimal LA and ALA and abundant DHA and ARA, for treatment of intestinal failure-associated liver disease, does not result in EFAD. These findings challenge the essentiality of LA and ALA in the presence of ARA and DHA. Evidence discussed in this review supports the idea that ARA and DHA can independently fulfill dietary essential fatty acid requirements. The imminent introduction of new ILE rich in ARA and DHA in the United States highlights the importance of understanding their essentiality, especially when provision of ALA and LA is below the established daily minimum requirement.


Subject(s)
Fat Emulsions, Intravenous/administration & dosage , Fatty Acids, Essential/administration & dosage , Animals , Arachidonic Acid/administration & dosage , Arachidonic Acid/biosynthesis , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/biosynthesis , Fatty Acids, Essential/deficiency , Female , Fish Oils/administration & dosage , Humans , Linoleic Acid/administration & dosage , Linoleic Acid/metabolism , Mice , Nutritional Requirements , Rats , Triglycerides , United States , alpha-Linolenic Acid/administration & dosage , alpha-Linolenic Acid/metabolism
18.
J Surg Res ; 212: 146-152, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28550900

ABSTRACT

BACKGROUND: Although commonly performed in adult swine, unilateral pneumonectomy in piglets requires significant modifications in the surgical approach and perioperative care because of their smaller size and limited physiological reserve. METHODS: Nineteen neonatal piglets underwent a left pneumonectomy. They were allowed 5-7 d of preoperative acclimation and nutritional optimization. Preoperative weight gain and laboratory values were obtained before the time of surgery. A "ventro-cranial" approach is adopted where components of the pulmonary hilum were sequentially identified and ligated, starting from the most ventral and cranial structure, the superior pulmonary vein. The principle of gentle ventilation was followed throughout the entire operation. RESULTS: The median age of the piglets at the time of surgery was 12 (10-12) d. The median preoperative weight gain and albumin level were 20% (16-26%) and 2.3 (2.1-2.4) g/dL, respectively. The median operative time was 59 (50-70) min. Five of the first nine piglets died from complications, two from poor preoperative nutritional optimization (both with <10% weight gain and 2 g/dL for albumin), one from an intubation complication, one from intra-operative bleeding, and one in the postoperative period from a ruptured bulla. No mortality occurred for the next 10 cases. CONCLUSIONS: Successful outcomes for unilateral pneumonectomy in piglets require special attention to preoperative nutritional optimization, gentle ventilation, and meticulous surgical dissection. Preoperative weight gain and albumin levels should be used to identify appropriate surgical candidates. The "ventro-cranial" approach allows for a technically straightforward completion of the procedure.


Subject(s)
Models, Animal , Perioperative Care/methods , Pneumonectomy/methods , Swine/surgery , Animals , Female
19.
J Cyst Fibros ; 16(4): 519-524, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28185886

ABSTRACT

BACKGROUND: Adequate nutrition is essential for growth in children with cystic fibrosis (CF). The new CF Foundation Clinical Practice Guidelines bring attention to monitoring macronutrient intake as well as total energy. METHODS: Dietary intake of 75 preschool children with CF and pancreatic insufficiency was examined and compared to the Clinical Practice Guidelines. Regression analyses examined relationships between macronutrient intake and growth. RESULTS: Approximately 45% of children met the 110% minimum recommended dietary allowance (RDA) recommendation. Children consumed 35.3% (6.1) of total daily energy intake from fat, 12.7% (1.7) from protein, and 52.0% (6.1) from carbohydrates. Percent energy from protein was associated with height growth. CONCLUSIONS: Many preschoolers with CF are not meeting nutrition benchmarks for total energy and fat. To optimize nutrition early, dietary monitoring with frequent individualized feedback is needed. Optimizing intake of macronutrients that promote growth, especially fat and protein, should be a primary clinical target.


Subject(s)
Cystic Fibrosis , Dietary Carbohydrates/analysis , Dietary Fats/analysis , Dietary Proteins/analysis , Exocrine Pancreatic Insufficiency , Recommended Dietary Allowances , Child Development/physiology , Child, Preschool , Cystic Fibrosis/diagnosis , Cystic Fibrosis/diet therapy , Cystic Fibrosis/epidemiology , Cystic Fibrosis/physiopathology , Diet Therapy/methods , Diet Therapy/standards , Energy Intake , Exocrine Pancreatic Insufficiency/diagnosis , Exocrine Pancreatic Insufficiency/epidemiology , Female , Growth/physiology , Humans , Male , Nutritional Status , Practice Guidelines as Topic , United States/epidemiology
20.
JPEN J Parenter Enteral Nutr ; 41(2): 181-187, 2017 02.
Article in English | MEDLINE | ID: mdl-26993989

ABSTRACT

BACKGROUND: Fat emulsions are important components of parenteral nutrition (PN). Fish oil (FO) emulsions reverse cholestasis in PN-associated liver disease. There are 2 FO monographs. One is "FO; rich in omega-3 fatty acids" (NFO). The other, "omega-3 acids," (PFO), is enriched in omega-3 fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). The purpose of this study is to compare the effects of 20% NFO and PFO emulsions produced in the laboratory in a murine model. METHODS: Emulsions were compounded containing different oils: soybean oil (SO), NFO, and two PFOs differing in percentage of fatty acids as triglycerides (PFO66 and PFO90). Chow-fed mice received saline, one of the above emulsions, or a commercial FO (OM) intravenously (2.4 g/kg/day) for 19 days. On day 19, animals were euthanized. Livers, spleens, and lungs were procured for histologic analysis. RESULTS: OM, SO, NFO, and PFO90 were well-tolerated clinically. PFO66 resulted in tachypnea and lethargy for ~1 minute following injections. At euthanasia, PFO66 and PFO90 groups had organomegaly. Histologically, these groups had splenic and hepatic fat-laden macrophages, and lungs had scattered fat deposits. Other groups had normal organs. CONCLUSIONS: PFO emulsions present an attractive possibility for improving inflammation in PN-dependent patients by concentrating anti-inflammatory EPA and DHA. However, 20% PFO emulsions were poorly tolerated and precipitated adverse end organ sequelae, suggesting that they may not be safe. Development of novel manufacturing methods may achieve safe 20% PFO parenteral emulsions, but by established formulation methods, these emulsions were clinically suboptimal despite meeting pharmacopeial standards.


Subject(s)
Fat Emulsions, Intravenous/chemistry , Fish Oils/chemistry , Parenteral Nutrition , Animals , Docosahexaenoic Acids/analysis , Eicosapentaenoic Acid/analysis , Fat Emulsions, Intravenous/administration & dosage , Fat Emulsions, Intravenous/adverse effects , Fatty Acids, Omega-3/analysis , Fish Oils/administration & dosage , Liver/drug effects , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Parenteral Nutrition/adverse effects , Soybean Oil , Spleen/drug effects , Triglycerides
SELECTION OF CITATIONS
SEARCH DETAIL
...