Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Inflammation ; 42(3): 946-952, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30535619

ABSTRACT

Effect of aging and hyperglycemia on oxidative stress (OS) and inflammation in dyslipidemic conditions has not been elucidated. Hence, in this study, we assessed the implications of aging, hyperglycemia, and also the dietary effect of n-3 fatty acids (α-linolenic acid (ALA) and eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA)) on OS and inflammation in dyslipidemic rats. Dyslipidemia was induced in young and aged rats by feeding high-fat lard (HFL) diet. Diabetes was induced in young dyslipidemic rats by administering streptozotocin 30 days after the induction of dyslipidemia. Experimental groups received diets containing canola oil (HF + CNO) and fish oil (HF + FO) as a source of ALA and EPA + DHA respectively. After 60 days of feeding rats with their respective diets, OS and inflammatory markers in serum were assessed. Dyslipidemia caused significant (p < 0.05) increase in OS (lipid peroxidation, nitric oxide, and protein carbonyl), pro-inflammatory cytokine (CRP, IL-1ß, MCP-1, and TNF-α), and eicosanoid (PGE2, LTB4, and LTC4) level in serum of both young and aged rats. Aged dyslipidemic rats presented significantly (p < 0.05) higher level of these markers compared to young dyslipidemic rats. Hyperglycemia onset further augmented OS and inflammatory markers in young dyslipidemic rats significantly (p < 0.05). Administration of n-3 fatty acids downregulated the serum markers of OS and inflammation in all the three experimental models. Thus, aging and hyperglycemia onset intensified dyslipidemia-induced OS and inflammation. Dietary preformed EPA + DHA presented larger restorative potentials than precursor ALA in countering OS and inflammation in all the three experimental models.


Subject(s)
Aging , Dyslipidemias/complications , Fatty Acids, Omega-3/therapeutic use , Hyperglycemia/congenital , Inflammation/drug therapy , Oxidative Stress/drug effects , Animals , Biomarkers/blood , Diabetes Mellitus, Experimental/chemically induced , Docosahexaenoic Acids , Eicosapentaenoic Acid , Inflammation/etiology , Rats , alpha-Linolenic Acid
2.
Diabetes Care ; 41(8): 1821-1828, 2018 08.
Article in English | MEDLINE | ID: mdl-30030258

ABSTRACT

Despite significant reductions in serious adverse perinatal outcomes for women with type 1 diabetes in pregnancy, the opposite effect has been observed for fetal overgrowth and associated complications, such as neonatal hypoglycemia, shoulder dystocia, and admission to the neonatal intensive care unit. In addition, infants born large for gestational age (LGA) have an increased lifetime risk of obesity, diabetes, and chronic disease. Although exposure to hyperglycemia plays an important role, women who seemingly achieve adequate glycemic control in pregnancy continue to experience a greater risk of excess fetal growth, leading to LGA neonates and macrosomia. We review potential contributors to excess fetal growth in pregnancies complicated by type 1 diabetes. In addition to hyperglycemia, we explore the role of glycemic variability, prepregnancy overweight and obesity, gestational weight gain, and maternal lipid levels. Greater understanding of the stimuli that drive excess fetal growth could lead to targeted management strategies in pregnant women with type 1 diabetes, potentially reducing the incidence of LGA neonates and the inherent risk of acute and long-term complications.


Subject(s)
Diabetes Mellitus, Type 1/complications , Fetal Macrosomia/etiology , Hyperglycemia/etiology , Pregnancy in Diabetics , Birth Weight/physiology , Diabetes Mellitus, Type 1/epidemiology , Female , Fetal Macrosomia/epidemiology , Gestational Age , Humans , Hyperglycemia/complications , Hyperglycemia/congenital , Hyperglycemia/epidemiology , Infant, Newborn , Infant, Newborn, Diseases/epidemiology , Infant, Newborn, Diseases/etiology , Pregnancy , Pregnancy Outcome/epidemiology , Pregnancy in Diabetics/epidemiology , Risk Factors
3.
Clin Perinatol ; 45(1): 41-59, 2018 03.
Article in English | MEDLINE | ID: mdl-29406006

ABSTRACT

Neonatal diabetes mellitus is likely to be due to an underlying monogenic defect when it occurs at less than 6 months of age. Early recognition and urgent genetic testing are important for predicting the clinical course and raising awareness of possible additional features. Early treatment of sulfonylurea-responsive types of neonatal diabetes may improve neurologic outcomes. It is important to distinguish neonatal diabetes mellitus from other causes of hyperglycemia in newborns. Other causes include infection, stress, inadequate pancreatic insulin production in preterm infants, among others. This review explores the diagnostic approach, mutation types, management, and clinical course of neonatal diabetes.


Subject(s)
Diabetes Mellitus/congenital , Diabetes Mellitus/diagnosis , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Humans , Hyperglycemia/congenital , Hypoglycemic Agents/therapeutic use , Infant, Extremely Premature , Infant, Newborn , Infant, Premature , Insulin/therapeutic use , Mutation , Sulfonylurea Compounds/therapeutic use
4.
Diabet Med ; 35(2): 173-183, 2018 02.
Article in English | MEDLINE | ID: mdl-29117445

ABSTRACT

AIMS: To examine whether, in neonates of mothers with Type 1, Type 2 and gestational diabetes, in-target intrapartum glycaemic control was associated with a lower risk of neonatal hypoglycaemia compared with out-of-target glycaemic control. METHODS: We searched PubMed and EMBASE for all available publications, regardless of year, based on a published protocol (PROSPERO CRD42016052439). Studies were excluded if they did not report original data or were animal studies. Data were extracted from published reports in duplicate using a prespecified data extraction form. The main outcome of interest was the association between in-target intrapartum glycaemic control and neonatal hypoglycaemia. RESULTS: We screened 2846 records for potential study inclusion; 23 studies, including approximately 2835 women with diabetes, were included in the systematic review. Only two of those studies specifically examined in-target vs out-of-target intrapartum glycaemic control. Of the studies included, six showed a relationship between intrapartum glucose and neonatal hypoglycaemia, five others showed a relationship in at least one of the analyses performed and 12 did not find a significant relationship. Only one study was identified as having a low risk of bias. CONCLUSIONS: There is a paucity of high-quality data supporting the association of glucose during labour and delivery with neonatal hypoglycaemia in pregnancies complicated by diabetes. Further studies are required to examine the impact of tight glycaemic targets in labour.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 2/prevention & control , Diabetes, Gestational/prevention & control , Hyperglycemia/congenital , Pregnancy in Diabetics/prevention & control , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Diabetes, Gestational/blood , Female , Humans , Hyperglycemia/blood , Hyperglycemia/prevention & control , Infant, Newborn , Pregnancy , Pregnancy in Diabetics/blood , Prenatal Care , Risk Factors
5.
Endocrinology ; 158(5): 1140-1151, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28324053

ABSTRACT

Premature infants have altered glucose regulation early in life and increased risk for diabetes in adulthood. Although prematurity leads to an increased risk of diabetes and metabolic syndrome in adult life, the role of hepatic glucose regulation and adaptation to an early extrauterine environment in preterm infants remain unknown. The purpose of this study was to investigate developmental differences in glucose metabolism, hepatic protein content, and gene expression of key insulin-signaling/gluconeogenic molecules. Fetal baboons were delivered at 67%, 75%, and term gestational age and euthanized at birth. Neonatal baboons were delivered prematurely (67% gestation), survived for two weeks, and compared with similar postnatal term animals and underwent serial hyperinsulinemic-euglycemic clamp studies. Premature baboons had decreased endogenous glucose production (EGP) compared with term animals. Consistent with these results, the gluconeogenic molecule, phosphoenolpyruvate carboxykinase messenger RNA, was decreased in preterm baboons compared with terms. Hepatic insulin signaling was altered by preterm birth as evidenced by decreased insulin receptor-ß, p85 subunit of phosphoinositide 3-kinase, phosphorylated insulin receptor substrate 1, and Akt-1 under insulin-stimulated conditions. Furthermore, preterm baboons failed to have the normal increase in glycogen synthase kinase-α from fetal to postnatal life. The blunted responses in hepatic insulin signaling may contribute to the hyperglycemia of prematurity, while impaired EGP leads to hypoglycemia of prematurity.


Subject(s)
Gluconeogenesis/physiology , Hyperglycemia/congenital , Insulin Resistance , Liver/metabolism , Premature Birth/metabolism , Animals , Animals, Newborn , Female , Glucose/metabolism , Hyperglycemia/etiology , Hyperglycemia/metabolism , Insulin/metabolism , Male , Metabolic Networks and Pathways , Papio , Pregnancy , Term Birth/metabolism
6.
J Clin Endocrinol Metab ; 102(4): 1141-1150, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28204515

ABSTRACT

Context: Offspring of women with gestational diabetes (O-GDM) or type 1 diabetes mellitus (O-T1DM) have been exposed to hyperglycemia in utero and have an increased risk of developing metabolic disease in adulthood. Design: In total, we recruited 206 adult offspring comprising the two fetal hyperglycemic groups, O-GDM and O-T1DM, and, as a control group, offspring from the background population (O-BP). Subcutaneous fat biopsies were obtained and preadipocyte cell cultures were established from adult male O-GDM (n = 18, age 30.1 ± 2.5 years), O-T1DM (n = 18, age 31.6 ± 2.2 years), and O-BP (n = 16; age, 31.5 ± 2.7 years) and cultured in vitro. Main Outcome Measures: First, we studied in vivo adipocyte histology. Second, we studied in vitro preadipocyte leptin secretion, gene expression, and LEP DNA methylation. This was studied in combination with in vitro preadipocyte lipogenesis, lipolysis, and mitochondrial respiration. Results: We show that subcutaneous adipocytes from O-GDM are enlarged compared with O-BP adipocytes. Preadipocytes isolated from male O-GDM and O-T1DM and cultured in vitro displayed decreased LEP promoter methylation, increased leptin gene expression, and elevated leptin secretion throughout differentiation, compared with adipocytes established from male O-BP. In addition, the preadipocytes demonstrated functional defects including decreased maximal mitochondrial capacity with increased lipolysis and decreased ability to store fatty acids when challenged with 3 days of extra fatty acid supply. Conclusions: Taken together, these findings show that intrinsic epigenetic and functional changes exist in preadipocyte cultures from individuals exposed to fetal hyperglycemia who are at increased risk of developing metabolic disease.


Subject(s)
Adipocytes/physiology , Adult Children , Diabetes Mellitus, Type 1 , Diabetes, Gestational , Hyperglycemia/congenital , Pregnancy in Diabetics , Prenatal Exposure Delayed Effects/physiopathology , Adult , Case-Control Studies , Cells, Cultured , Diabetes Mellitus, Type 2/etiology , Disease Susceptibility , Female , Fetal Diseases/metabolism , Fetal Diseases/physiopathology , Humans , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Lipolysis/physiology , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Risk Factors
7.
Early Hum Dev ; 91(11): 655-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26385449

ABSTRACT

Preterm hyperglycaemia in the first 2 weeks of life is common under 29 weeks gestation and is associated with increased mortality and morbidity. While the definition of hyperglycaemia is reasonably consistent (>8 mmol/L) the treatment threshold varies widely in clinical practice. Insulin therapy is the most common approach despite international guidance urging caution because of hypoglycaemia. Significant hypoglycaemia is unusual outside studies targeting normoglycaemia. Insulin treatment also forms part of a nutritional strategy aiming to optimise early protein and energy intake so minimising the risk of preterm postnatal growth failure. Early parenteral amino acids also improve blood glucose control. There is some evidence of improved postnatal head growth with this approach but longer term neurodevelopmental studies are required. Glucose reduction is the alternative approach. This compromises early nutritional intake but avoids the potential for long-term cardiovascular and metabolic complications linked with high postnatal nutritional intakes and theoretically, insulin treatment.


Subject(s)
Hyperglycemia/drug therapy , Infant, Premature, Diseases/drug therapy , Insulin/therapeutic use , Amino Acids/therapeutic use , Blood Glucose/drug effects , Blood Glucose/metabolism , Gestational Age , Humans , Hyperglycemia/congenital , Hyperglycemia/diet therapy , Hyperglycemia/epidemiology , Infant Nutritional Physiological Phenomena , Infant, Newborn , Infant, Premature/blood , Infant, Premature, Diseases/epidemiology , Insulin/adverse effects , Parenteral Nutrition/methods , Risk Assessment
8.
Horm Res Paediatr ; 81(1): 43-9, 2014.
Article in English | MEDLINE | ID: mdl-24281139

ABSTRACT

BACKGROUND/AIMS: Term small-for-gestational-age and preterm born infants have an increased prevalence of metabolic syndrome components already in childhood. Data in very-low-birth-weight (VLBW) children are limited. We investigated the prevalence of metabolic syndrome components in VLBW infants at 2 years of corrected age. METHODS: We included 38 children, participating in the Neonatal Insulin Replacement Therapy in Europe (NIRTURE) trial, a randomized controlled trial of early insulin therapy in VLBW infants. Metabolic syndrome components were defined as: body mass index SDS >2; blood pressure (systolic and/or diastolic) ≥ 90th percentile; triglycerides ≥ 0.98 mmol/l; high-density lipoprotein (HDL) cholesterol ≤ 1.03 mmol/l; glucose ≥ 5.6 mmol/l. RESULTS: Two children (5%) had three metabolic syndrome components, 13 children (34%) had two components, and 11 children (29%) one component. 63% had raised blood pressure (prevalence higher in boys), 32% low HDL, and 30% high triglycerides (prevalence lower in early insulin group). In children with body mass index SDS <0, insulin-treated children had higher HDL than children with standard care. Systolic blood pressure was correlated with growth between term and 2 years of corrected age. CONCLUSIONS: VLBW infants already have a high prevalence of metabolic syndrome components at 2 years of corrected age. Early insulin treatment could have long-term benefits for some of these components.


Subject(s)
Infant, Very Low Birth Weight/metabolism , Metabolic Syndrome/epidemiology , Metabolic Syndrome/etiology , Adult , Age of Onset , Body Mass Index , Child, Preschool , Europe , Female , Humans , Hyperglycemia/congenital , Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Infant, Newborn , Infant, Premature, Diseases/drug therapy , Infant, Very Low Birth Weight/growth & development , Insulin/therapeutic use , Male , Standard of Care
9.
Nutrition ; 29(4): 688-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23466053

ABSTRACT

OBJECTIVE: Newborns of diabetic mothers have abnormal circulatory organs, so in this study, we explore insulin signaling in the newborn rat heart. METHODS: Pregnant rats were divided into streptozotocin-induced diabetic groups (DM) and control groups (CM). Rats were fed lard (21% fat), fish oil (21% fat), or a control diet (7% fat). To examine changes in insulin signaling in the hearts of infants of diabetic mothers (IDM) in relation to diet, we isolated the hearts from the IDM and control infants and determined the phosphorylation levels of Akt308, Akt473, p38, c-jun-NH2-terminal protein kinase (JNK), and extracellular signal-regulated protein kinase (ERK), and the expression levels of phosphoinositide-dependent protein kainase1 (PDK1) and mammalian target of rapamycin (mTOR). RESULTS: The mean blood glucose levels in the DM group and their infants were significantly higher than those in the CM group (P < 0.05) and their infants (P < 0.05), but the mean blood glucose levels of all infants was normal on postnatal d 4. Phosphorylation levels of Akt (Thr 308) (P < 0.05) and Akt (Ser 473) and the expression levels of PDK1 and mTOR were lower in infants of diabetic mothers (IDM) than in control infants. The phosphorylation level of Akt (Ser 473) and the expression level of mTOR increased in IDM fed the fish oil diet compared with those fed the lard diet (P < 0.05). CONCLUSION: A maternal diet rich in fish oil improves cardiac Akt-related signaling in the offspring of diabetic rats.


Subject(s)
Fish Oils/therapeutic use , MAP Kinase Signaling System , Maternal Nutritional Physiological Phenomena , Myometrium/metabolism , Prediabetic State/prevention & control , Pregnancy in Diabetics/diet therapy , Proto-Oncogene Proteins c-akt/metabolism , Animals , Animals, Newborn , Diet, High-Fat/adverse effects , Female , Fish Oils/adverse effects , Hyperglycemia/congenital , Hyperglycemia/prevention & control , Hypertriglyceridemia/complications , Hypertriglyceridemia/congenital , Hypertriglyceridemia/prevention & control , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Phosphorylation , Prediabetic State/complications , Prediabetic State/congenital , Prediabetic State/metabolism , Pregnancy , Pregnancy in Diabetics/blood , Protein Processing, Post-Translational , Rats , Serine/metabolism , Threonine/metabolism
10.
J Clin Invest ; 122(10): 3516-28, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23006325

ABSTRACT

Pancreatic agenesis is a human disorder caused by defects in pancreas development. To date, only a few genes have been linked to pancreatic agenesis in humans, with mutations in pancreatic and duodenal homeobox 1 (PDX1) and pancreas-specific transcription factor 1a (PTF1A) reported in only 5 families with described cases. Recently, mutations in GATA6 have been identified in a large percentage of human cases, and a GATA4 mutant allele has been implicated in a single case. In the mouse, Gata4 and Gata6 are expressed in several endoderm-derived tissues, including the pancreas. To analyze the functions of GATA4 and/or GATA6 during mouse pancreatic development, we generated pancreas-specific deletions of Gata4 and Gata6. Surprisingly, loss of either Gata4 or Gata6 in the pancreas resulted in only mild pancreatic defects, which resolved postnatally. However, simultaneous deletion of both Gata4 and Gata6 in the pancreas caused severe pancreatic agenesis due to disruption of pancreatic progenitor cell proliferation, defects in branching morphogenesis, and a subsequent failure to induce the differentiation of progenitor cells expressing carboxypeptidase A1 (CPA1) and neurogenin 3 (NEUROG3). These studies address the conserved and nonconserved mechanisms underlying GATA4 and GATA6 function during pancreas development and provide a new mouse model to characterize the underlying developmental defects associated with pancreatic agenesis.


Subject(s)
GATA4 Transcription Factor/physiology , GATA6 Transcription Factor/physiology , Gene Expression Regulation, Developmental/genetics , Organogenesis/genetics , Pancreas/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Binding Sites , Carboxypeptidases A/analysis , Cell Differentiation , Cell Division , Cell Lineage , Disease Models, Animal , Endoderm/metabolism , Epithelial Cells/pathology , GATA4 Transcription Factor/deficiency , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/deficiency , GATA6 Transcription Factor/genetics , Gene Knockdown Techniques , Genotype , Gestational Age , Hyperglycemia/congenital , Hyperglycemia/genetics , Insulin/metabolism , Insulin Secretion , Mice , Nerve Tissue Proteins/analysis , Organ Specificity , Pancreas/abnormalities , Pancreas/pathology , Transcription, Genetic
11.
J Clin Invest ; 122(10): 3504-15, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23006330

ABSTRACT

Recently, heterozygous mutations in GATA6 have been found in neonatal diabetic patients with failed pancreatic organogenesis. To investigate the roles of GATA4 and GATA6 in mouse pancreas organogenesis, we conditionally inactivated these genes within the pancreas. Single inactivation of either gene did not have a major impact on pancreas formation, indicating functional redundancy. However, double Gata4/Gata6 mutant mice failed to develop pancreata, died shortly after birth, and displayed hyperglycemia. Morphological defects in Gata4/Gata6 mutant pancreata were apparent during embryonic development, and the epithelium failed to expand as a result of defects in cell proliferation and differentiation. The number of multipotent pancreatic progenitors, including PDX1+ cells, was reduced in the Gata4/Gata6 mutant pancreatic epithelium. Remarkably, deletion of only 1 Gata6 allele on a Gata4 conditional knockout background severely reduced pancreatic mass. In contrast, a single WT allele of Gata4 in Gata6 conditional knockout mice was sufficient for normal pancreatic development, indicating differential contributions of GATA factors to pancreas formation. Our results place GATA factors at the top of the transcriptional network hierarchy controlling pancreas organogenesis.


Subject(s)
GATA4 Transcription Factor/physiology , GATA6 Transcription Factor/physiology , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Organogenesis/genetics , Pancreas/embryology , Trans-Activators/genetics , Animals , Binding Sites , Cell Differentiation , Cell Division , Epithelial Cells/pathology , GATA4 Transcription Factor/deficiency , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/deficiency , GATA6 Transcription Factor/genetics , Genotype , Homeodomain Proteins/biosynthesis , Hyperglycemia/congenital , Hyperglycemia/genetics , Mice , Mice, Knockout , Mice, Transgenic , Pancreas/abnormalities , Pancreas/pathology , Promoter Regions, Genetic/genetics , Protein Binding , Trans-Activators/biosynthesis , Transcription, Genetic
12.
J Matern Fetal Neonatal Med ; 25(11): 2415-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22668010

ABSTRACT

BACKGROUND/OBJECTIVE: Hyperglycaemia is common in very premature neonates and is associated with increased risk of intraventricular haemorrhage, necrotising enterocolitis, sepsis and death. Administration of insulin may risk hypoglycaemia and associated complications. To determine effects of insulin infusions in very premature infants on morbidity, mortality and long-term neurodevelopmental outcome. METHODS: Retrospective audit of 97 infants delivered at <29 weeks gestation and admitted to The Canberra Hospital NICU. Data on insulin treatment, Blood Glucose Levels (BGL's) prior and during insulin therapy, episodes of significant hypoglycaemia and neurodevelopmental outcome at 12 months corrected age was collected. RESULTS: 17 (17.5%) neonates received insulin. Episodes of hypoglycaemia were infrequent (1.3%, 95% CI 0.5-2.9). Multiple regression analysis showed that insulin treatment was not associated with an increased risk of retinopathy of prematurity (OR 3.6, 95% CI 0.4-32.3) or mortality (OR 1.2, 95% CI 0.29-5.0). No significant difference in 12 month neurodevelopmental or anthropometric outcomes was detected in infants who received insulin. CONCLUSION: Insulin infusions for hyperglycaemia appear to be safe with infrequent episodes of hypoglycaemia, no increased risk of morbidity or mortality and no adverse effect on long-term neurodevelopmental outcome.


Subject(s)
Brain/growth & development , Child Development/drug effects , Hyperglycemia/drug therapy , Infant, Extremely Premature , Infant, Premature, Diseases/drug therapy , Insulin/administration & dosage , Brain/drug effects , Child Development/physiology , Female , Humans , Hyperglycemia/congenital , Hyperglycemia/epidemiology , Hyperglycemia/mortality , Infant, Extremely Premature/growth & development , Infant, Extremely Premature/metabolism , Infant, Extremely Premature/physiology , Infant, Newborn , Infant, Premature, Diseases/epidemiology , Infant, Premature, Diseases/mortality , Insulin/adverse effects , Insulin Infusion Systems , Male , Morbidity , Retrospective Studies , Survival Analysis , Time Factors , Treatment Outcome
13.
Early Hum Dev ; 87 Suppl 1: S19-22, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21276670

ABSTRACT

Neonatal hyperglycaemia is a frequent complication in VLBW infants during the first week of life. The more common causes include high glucose intake, stress situations such as sepsis, NEC, and surgical treatments, as well as the administration of vasoactive drugs and methylxanthines. The appropriate definition is unclear. Hyperglycaemia has been associated with increased mortality and major morbidities. There have been insufficient randomized clinical trials to help in clarifying which infants should be treated, and there are insufficient data on the pharmacokinetics of insulin in these vulnerable patients.


Subject(s)
Hyperglycemia/congenital , Hyperglycemia/therapy , Infant, Premature, Diseases/therapy , Infant, Premature , Diabetes Mellitus/congenital , Diabetes Mellitus/epidemiology , Diabetes Mellitus/therapy , Glucose Intolerance/congenital , Glucose Intolerance/etiology , Glucose Intolerance/therapy , Humans , Hyperglycemia/epidemiology , Hyperglycemia/etiology , Incidence , Infant, Newborn , Infant, Premature, Diseases/epidemiology , Infant, Premature, Diseases/etiology
14.
BMC Pediatr ; 11: 10, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21294901

ABSTRACT

BACKGROUND: To examine the blood glucose profile and the relationship between blood glucose levels and neurodevelopmental outcome in term infants with hypoxic-ischaemic encephalopathy. METHODS: Blood glucose values within 72 hours of birth were collected from 52 term infants with hypoxic-ischaemic encephalopathy. Hypoglycaemia [< 46.8 mg/dL (2.6 mmol/L)] and hyperglycaemia [> 150 mg/dL (8.3 mmol/L)] were correlated to neurodevelopmental outcome at 24 months of age. RESULTS: Four fifths of the 468 blood samples were in the normoglycaemic range (392/468:83.8%). Of the remaining 76 samples, 51.3% were in the hypoglycaemic range and (48.7%) were hyperglycaemic. A quarter of the hypoglycaemic samples (28.2%:11/39) and a third of the hyperglycaemic samples (32.4%:12/37) were recorded within the first 30 minutes of life. Mean (SD) blood glucose values did not differ between infants with normal and abnormal outcomes [4.89(2.28) mmol/L and 5.02(2.35) mmol/L, p value = 0.15] respectively. In term infants with hypoxic-ischaemic encephalopathy, early hypoglycaemia (between 0-6 hours of life) was associated with adverse outcome at 24 months of age [OR = 5.8, CI = 1.04-32)]. On multivariate analysis to adjust for grade of HIE this association was not statistically significant. Late hypoglycaemia (6-72 hours of life) was not associated with abnormal outcome [OR = 0.22, CI (0.04-1.14)]. The occurrence of hyperglycaemia was not associated with adverse outcome. CONCLUSION: During the first 72 hours of life, blood glucose profile in infants with hypoxic-ischaemic encephalopathy varies widely despite a management protocol. Early hypoglycaemia (0-6 hours of life) was associated with severe HIE, and thereby; adverse outcome.


Subject(s)
Asphyxia Neonatorum/blood , Blood Glucose/analysis , Brain Damage, Chronic/epidemiology , Hypoglycemia/epidemiology , Hypoxia-Ischemia, Brain/blood , Asphyxia Neonatorum/complications , Biomarkers , Birth Weight , Brain Damage, Chronic/etiology , Female , Follow-Up Studies , Gestational Age , Humans , Hyperglycemia/congenital , Hyperglycemia/epidemiology , Hypoglycemia/complications , Hypoglycemia/congenital , Hypothermia, Induced , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/therapy , Infant, Newborn , Male , Retrospective Studies , Time Factors , Treatment Outcome
15.
J Matern Fetal Neonatal Med ; 23(3): 199-203, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20121460

ABSTRACT

Gestational diabetes mellitus (GDM) is a type of diabetes that presents during pregnancy and usually disappears shortly after a woman gives birth. Better recognition of the risk factors of GDM, combined with more universal screening for the disease in many countries, has led to the increased detection of GDM along with other forms of pregestational diabetes. There is growing evidence that GDM significantly increases the risk of a number of short- and long-term adverse consequences for the fetus and mother, the most significant of which is a predisposition to the development of metabolic syndrome and Type 2 diabetes. Maternal and childhood obesity as well as cardiovascular disease are also potential long-term consequences of GDM. On the other hand, there is a growing body of evidence suggesting that the risk of many of these consequences can be significantly reduced or eliminated by aggressive treatment of GDM. There remains, however, a great deal of controversy over when to begin screening for hyperglycemia in pregnancy and at what level of hyperglycemia should aggressive intervention be initiated.


Subject(s)
Diabetes, Gestational/physiopathology , Fetal Diseases/etiology , Maternal-Fetal Exchange/physiology , Mothers , Diabetes, Gestational/epidemiology , Female , Fetal Diseases/epidemiology , Humans , Hyperglycemia/complications , Hyperglycemia/congenital , Hyperglycemia/epidemiology , Hyperglycemia/etiology , Infant, Newborn , Infant, Newborn, Diseases/epidemiology , Infant, Newborn, Diseases/etiology , Pregnancy , Pregnancy Complications/epidemiology , Pregnancy Complications/etiology , Risk Factors
17.
Diabet Med ; 25(4): 450-4, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18387079

ABSTRACT

AIMS: To assess a new technique for continuous monitoring of glucose concentration during labour in diabetic mothers. A second objective was to study maternal glucose levels in relation to postnatal glucose adaptation and the need for intravenous (IV) glucose treatment in the newborn infant. METHODS: Fifteen pregnant women with insulin-treated diabetes mellitus participated in this prospective pilot study. To measure their glucose control during labour we used the Continuous Subcutaneous Glucose Monitoring System (CGMS; Medtronic, Minneapolis, MN, USA) to calculate the mean glucose concentration and the area under the curve (AUC) in the last 120 min before delivery. All infants of these women were transferred to the neonatal care unit for early oral feeding and blood glucose measurements up to 14 h after delivery. Infants received IV glucose if blood glucose values were repeatedly < 2.2 mmol/l. RESULTS: All women coped well with the CGMS monitoring. AUC 0-120 min before delivery, mean glucose concentration 0-120 min before delivery and cord plasma insulin level were all significantly associated with the need for IV glucose in the newborn children. CONCLUSIONS: In this study we found an association between maternal glucose concentrations during labour and postnatal glucose adaptation and need for IV glucose treatment in the infants. Online monitoring of glucose levels during delivery might help us to achieve maternal normoglycaemia and further reduce the risk of postnatal hypoglycaemia in the offspring.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes, Gestational/metabolism , Hyperglycemia/congenital , Infant, Newborn/metabolism , Pregnancy in Diabetics/metabolism , Female , Humans , Monitoring, Physiologic/methods , Pregnancy , Prospective Studies , Treatment Outcome
20.
Neonatal Netw ; 19(4): 13-20, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11949098

ABSTRACT

Except for the hyperinsulinism associated with the infant of a diabetic mother (accounting for about 5 percent of NICU admissions annually), pancreatic disorders of the newborn are rare. Congenital anomalies (such as annular pancreas) and endocrine disorders (such as hyperinsulinism of nesidioblastosis or hyperglycemia of neonatal diabetes mellitus) present many challenges to the personnel caring for these infants and their families. The potential mortality and morbidity of these disorders make it imperative for nurses and nurse practitioners working with infants to recognize and understand pancreatic dysfunction so that appropriate and timely intervention can prevent complications of brain injury and developmental delay. The home care needs of these infants and the extensive teaching needs of their parents require skilled nursing care to ensure a safe discharge.


Subject(s)
Digestive System Abnormalities/diagnosis , Pancreatic Diseases/congenital , Pancreatic Diseases/diagnosis , Diabetes Mellitus/congenital , Diabetes Mellitus/diagnosis , Diabetes Mellitus/nursing , Digestive System Abnormalities/nursing , Female , Humans , Hyperglycemia/congenital , Hyperglycemia/diagnosis , Hyperglycemia/nursing , Hyperinsulinism/congenital , Hyperinsulinism/diagnosis , Hyperinsulinism/nursing , Incidence , Infant, Newborn , Male , Pancreatic Diseases/nursing , Prognosis , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...