Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 462
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(6): 2914-2922, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31974308

RESUMO

The epidermis-specific lipid acylceramide plays a pivotal role in the formation of the permeability barrier in the skin; abrogation of its synthesis causes the skin disorder ichthyosis. However, the acylceramide synthetic pathway has not yet been fully elucidated: Namely, the acyl-CoA synthetase (ACS) involved in this pathway remains to be identified. Here, we hypothesized it to be encoded by FATP4/ACSVL4, the causative gene of ichthyosis prematurity syndrome (IPS). In vitro experiments revealed that FATP4 exhibits ACS activity toward an ω-hydroxy fatty acid (FA), an intermediate of the acylceramide synthetic pathway. Fatp4 knockout (KO) mice exhibited severe skin barrier dysfunction and morphological abnormalities in the epidermis. The total amount of acylceramide in Fatp4 KO mice was reduced to ∼10% of wild-type mice. Decreased levels and shortening of chain lengths were observed in the saturated, nonacylated ceramides. FA levels were not decreased in the epidermis of Fatp4 KO mice. The expression levels of the FA elongase Elovl1 were reduced in Fatp4 KO epidermis, partly accounting for the reduction and shortening of saturated, nonacylated ceramides. A decrease in acylceramide levels was also observed in human keratinocytes with FATP4 knockdown. From these results, we conclude that skin barrier dysfunction observed in IPS patients and Fatp4 KO mice is caused mainly by reduced acylceramide production. Our findings further elucidate the molecular mechanism governing acylceramide synthesis and IPS pathology.


Assuntos
Ceramidas/metabolismo , Epiderme/metabolismo , Proteínas de Transporte de Ácido Graxo/metabolismo , Ictiose/metabolismo , Doenças do Prematuro/metabolismo , Animais , Ceramidas/química , Proteínas de Transporte de Ácido Graxo/genética , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Feminino , Humanos , Ictiose/genética , Doenças do Prematuro/genética , Masculino , Camundongos Knockout , Permeabilidade , Pele/metabolismo
2.
Pediatr Res ; 89(6): 1405-1413, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33003189

RESUMO

BACKGROUND: Identifying preterm infants at risk for mortality or major morbidity traditionally relies on gestational age, birth weight, and other clinical characteristics that offer underwhelming utility. We sought to determine whether a newborn metabolic vulnerability profile at birth can be used to evaluate risk for neonatal mortality and major morbidity in preterm infants. METHODS: This was a population-based retrospective cohort study of preterm infants born between 2005 and 2011 in California. We created a newborn metabolic vulnerability profile wherein maternal/infant characteristics along with routine newborn screening metabolites were evaluated for their association with neonatal mortality or major morbidity. RESULTS: Nine thousand six hundred and thirty-nine (9.2%) preterm infants experienced mortality or at least one complication. Six characteristics and 19 metabolites were included in the final metabolic vulnerability model. The model demonstrated exceptional performance for the composite outcome of mortality or any major morbidity (AUC 0.923 (95% CI: 0.917-0.929). Performance was maintained across mortality and morbidity subgroups (AUCs 0.893-0.979). CONCLUSIONS: Metabolites measured as part of routine newborn screening can be used to create a metabolic vulnerability profile. These findings lay the foundation for targeted clinical monitoring and further investigation of biological pathways that may increase the risk of neonatal death or major complications in infants born preterm. IMPACT: We built a newborn metabolic vulnerability profile that could identify preterm infants at risk for major morbidity and mortality. Identifying high-risk infants by this method is novel to the field and outperforms models currently in use that rely primarily on infant characteristics. Utilizing the newborn metabolic vulnerability profile for precision clinical monitoring and targeted investigation of etiologic pathways could lead to reductions in the incidence and severity of major morbidities associated with preterm birth.


Assuntos
Mortalidade Infantil , Recém-Nascido Prematuro , Morbidade , Adulto , Feminino , Humanos , Lactente , Recém-Nascido , Doenças do Prematuro/metabolismo , Doenças do Prematuro/mortalidade , Gravidez , Fatores de Risco , Adulto Jovem
3.
Nature ; 506(7487): 230-4, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24390343

RESUMO

There are no clinically relevant treatments available that improve function in the growing population of very preterm infants (less than 32 weeks' gestation) with neonatal brain injury. Diffuse white matter injury (DWMI) is a common finding in these children and results in chronic neurodevelopmental impairments. As shown recently, failure in oligodendrocyte progenitor cell maturation contributes to DWMI. We demonstrated previously that the epidermal growth factor receptor (EGFR) has an important role in oligodendrocyte development. Here we examine whether enhanced EGFR signalling stimulates the endogenous response of EGFR-expressing progenitor cells during a critical period after brain injury, and promotes cellular and behavioural recovery in the developing brain. Using an established mouse model of very preterm brain injury, we demonstrate that selective overexpression of human EGFR in oligodendrocyte lineage cells or the administration of intranasal heparin-binding EGF immediately after injury decreases oligodendroglia death, enhances generation of new oligodendrocytes from progenitor cells and promotes functional recovery. Furthermore, these interventions diminish ultrastructural abnormalities and alleviate behavioural deficits on white-matter-specific paradigms. Inhibition of EGFR signalling with a molecularly targeted agent used for cancer therapy demonstrates that EGFR activation is an important contributor to oligodendrocyte regeneration and functional recovery after DWMI. Thus, our study provides direct evidence that targeting EGFR in oligodendrocyte progenitor cells at a specific time after injury is clinically feasible and potentially applicable to the treatment of premature children with white matter injury.


Assuntos
Lesões Encefálicas/congênito , Lesões Encefálicas/tratamento farmacológico , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/uso terapêutico , Oligodendroglia/efeitos dos fármacos , Administração Intranasal , Animais , Animais Recém-Nascidos , Lesões Encefálicas/patologia , Lesões Encefálicas/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doenças Desmielinizantes/congênito , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/prevenção & controle , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/administração & dosagem , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Hipóxia/patologia , Hipóxia/fisiopatologia , Doenças do Prematuro/tratamento farmacológico , Doenças do Prematuro/metabolismo , Doenças do Prematuro/patologia , Masculino , Camundongos , Terapia de Alvo Molecular , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Fatores de Tempo
4.
J Neurophysiol ; 122(5): 1874-1883, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31483699

RESUMO

In neonates, catecholamine (CA) secretion from adrenal medullary chromaffin cells (AMC) is an important mechanism for maintaining homeostasis during hypoxia. Nearly 90% of premature infants experience chronic intermittent hypoxia (IH) because of high incidence of apnea of prematurity, which is characterized by periodic stoppage of breathing. The present study examined the effects of repetitive hypoxia, designed to mimic apnea of prematurity, on CA release from AMC of neonatal rats. Neonatal rats were exposed to either control conditions or chronic intermittent hypoxia (IH) from ages postnatal days 0-5 (P0-P5), and CA release from adrenal medullary slices was measured after challenge with repetitive hypoxia (5 episodes of 30-s hypoxia, Po2 ~35 mmHg). In response to repetitive hypoxia, chronic IH-treated AMC exhibited sustained CA release, and this phenotype was not seen in control AMC. The sustained CA release was associated with long-lasting elevation of intracellular Ca2+ concentration ([Ca2+]i), which was due to store-operated Ca2+ entry (SOCE). 2-Aminoethoxydiphenyl borate, an inhibitor of SOCE, prevented the long-lasting [Ca2+]i elevation and CA release. Repetitive hypoxia increased H2O2 abundance, and polyethylene glycol (PEG)-catalase, a scavenger of H2O2 blocked this effect. PEG-catalase also prevented repetitive hypoxia-induced SOCE activation, sustained [Ca2+]i elevation, and CA release. These results demonstrate that repetitive hypoxia induces long-term facilitation of CA release in chronic IH-treated neonatal rat AMC through sustained Ca2+ influx mediated by SOCE.NEW & NOTEWORTHY Apnea of prematurity and the resulting chronic intermittent hypoxia are major clinical problems in neonates born preterm. Catecholamine release from adrenal medullary chromaffin cells maintains homeostasis during hypoxia in neonates. Our results demonstrate that chronic intermittent hypoxia induces a hitherto uncharacterized long-term facilitation of catecholamine secretion from neonatal rat chromaffin cells in response to repetitive hypoxia, simulating hypoxic episodes encountered during apnea of prematurity. The sustained catecholamine secretion might contribute to cardiovascular morbidities in infants with apnea of prematurity.


Assuntos
Medula Suprarrenal/metabolismo , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Hipóxia/metabolismo , Animais , Animais Recém-Nascidos , Apneia/metabolismo , Doença Crônica , Modelos Animais de Doenças , Doenças do Prematuro/metabolismo , Ratos , Ratos Sprague-Dawley
5.
Dig Dis Sci ; 64(11): 3078-3085, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30989465

RESUMO

Necrotizing enterocolitis (NEC) is one of the most severe diseases of preterm neonates and has a high mortality rate. With the development of inspection techniques and new biomarkers, the diagnostic accuracy of NEC is constantly improving. The most recognized potential risk factors include prematurity, formula-feeding, infection, and microbial dysbiosis. With further understanding of the pathogenesis, more effective prevention and therapies will be applied to clinical or experimental NEC. At present, such new potential prevention and therapies for NEC are mainly focused on the Toll-like receptor 4 inflammatory signaling pathway, the repair of intestinal barrier function, probiotics, antioxidative stress, breast-feeding, and immunomodulatory agents. Many new studies have changed our understanding of the pathogenesis of NEC and improve our approaches for preventing and treating of NEC each year. This review provides an overview of the recent researches focused on clinical or experimental NEC and highlights the advances made within the past 5 years toward the development of new potential preventive approaches and therapies for this disease.


Assuntos
Aleitamento Materno , Enterocolite Necrosante/prevenção & controle , Recém-Nascido de Baixo Peso , Doenças do Prematuro/prevenção & controle , Probióticos/uso terapêutico , Animais , Aleitamento Materno/tendências , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/terapia , Humanos , Recém-Nascido de Baixo Peso/metabolismo , Recém-Nascido , Doenças do Prematuro/metabolismo , Doenças do Prematuro/terapia , Receptor 4 Toll-Like/metabolismo
6.
J Pediatr ; 198: 194-200.e3, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29661562

RESUMO

OBJECTIVE: To evaluate the association between early metabolic profiles combined with infant characteristics and survival past 7 days of age in infants born at 22-25 weeks of gestation. STUDY DESIGN: This nested case-control consisted of 465 singleton live births in California from 2005 to 2011 at 22-25 weeks of gestation. All infants had newborn metabolic screening data available. Data included linked birth certificate and mother and infant hospital discharge records. Mortality was derived from linked death certificates and death discharge information. Each death within 7 days was matched to 4 surviving controls by gestational age and birth weight z score category, leaving 93 cases and 372 controls. The association between explanatory variables and 7-day survival was modeled via stepwise logistic regression. Infant characteristics, 42 metabolites, and 12 metabolite ratios were considered for model inclusion. Model performance was assessed via area under the curve. RESULTS: The final model included 1 characteristic and 11 metabolites. The model demonstrated a strong association between metabolic patterns and infant survival (area under the curve [AUC] 0.885, 95% CI 0.851-0.920). Furthermore, a model with just the selected metabolites performed better (AUC 0.879, 95% CI 0.841-0.916) than a model with multiple clinical characteristics (AUC 0.685, 95% CI 0.627-0.742). CONCLUSIONS: Use of metabolomics significantly strengthens the association with 7-day survival in infants born extremely premature. Physicians may be able to use metabolic profiles at birth to refine mortality risks and inform postnatal counseling for infants born at <26 weeks of gestation.


Assuntos
Doenças do Prematuro/metabolismo , Doenças do Prematuro/mortalidade , Metaboloma , California , Estudos de Casos e Controles , Idade Gestacional , Humanos , Lactente , Mortalidade Infantil , Recém-Nascido , Recém-Nascido Prematuro , Modelos Logísticos , Triagem Neonatal , Taxa de Sobrevida
7.
J Pediatr Gastroenterol Nutr ; 66(6): e153-e157, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29470288

RESUMO

OBJECTIVES: The aim of this study was to determine the effect of digested whole human milk (HM; first sample available after birth from mothers of premature infants) on inflammation, oxidative stress, and cytotoxicity in Caco-2 human intestinal epithelial cells stimulated with lipopolysaccharides or tumor necrosis factor (TNF) to mimic the potential in vivo insults facing the premature infant's gastrointestinal tract. METHODS: Fully differentiated Caco-2 cells were exposed to digested HM (n = 10; samples from 10 different individuals) before stimulation with lipopolysaccharides, TNF, or no stimulation overnight. Inflammation was determined by production of interleukin-8, oxidative stress by levels of F2-isoprostane, and cytotoxicity by released lactate dehydrogenase. RESULTS: HM significantly suppressed interleukin-8 production and cytotoxicity in TNF-stimulated cells, while also suppressing cell death under baseline conditions. Individual HM samples differed widely in their ability to modulate cellular responses. CONCLUSIONS: Results from this study provide evidence that digested HM can reduce both an exaggerated inflammatory response and intestinal damage that contribute to the pathogenesis of necrotizing enterocolitis.


Assuntos
Morte Celular/imunologia , Enterocolite Necrosante/prevenção & controle , Doenças do Prematuro/prevenção & controle , Recém-Nascido Prematuro/imunologia , Inflamação/prevenção & controle , Mucosa Intestinal/imunologia , Leite Humano/imunologia , Biomarcadores/metabolismo , Células CACO-2 , Enterocolite Necrosante/etiologia , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/metabolismo , Humanos , Recém-Nascido , Recém-Nascido Prematuro/metabolismo , Doenças do Prematuro/etiologia , Doenças do Prematuro/imunologia , Doenças do Prematuro/metabolismo , Inflamação/etiologia , Inflamação/imunologia , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Lipopolissacarídeos , Leite Humano/metabolismo , Estresse Oxidativo/imunologia , Fator de Necrose Tumoral alfa/metabolismo
8.
BMC Pediatr ; 18(1): 66, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29452581

RESUMO

BACKGROUND: What constitutes a hemodynamically relevant patent ductus arteriosus (hrPDA) in preterm infants is unclear. Different clinical and echocardiographic parameters are used, but a gold standard definition is lacking. Our objective was to evaluate associations between regional cerebral tissue oxygen saturation (rcStO2), fraction of tissue oxygen extraction (rcFtO2E) measured by frequency domain near-infrared spectroscopy (fd-NIRS) and their correlation to echocardiographic, Doppler-ultrasound, and clinical parameters in preterm infants with and without a hrPDA. METHODS: In this prospective observational study, 22 infants < 1500 g (mean [± SD]: gestational age 28.6 [±1.8] weeks, birth weight 1076 [±284] g, median (interquartile range) postnatal age at measurement 7.6 (4.6-12.9) d) with a clinical suspicion of hrPDA were analysed. Twelve infants had left-to-right shunt through PDA, and in 6 of these the PDA was classified as hrPDA based on pre-defined clinical and echocardiographic criteria. fd-NIRS, echocardiographic and Doppler-ultrasound examinations were performed. After identification of blood hemoglobin (Hb) as confounding factor, rcStO2 and rcFtO2E were corrected for this effect. RESULTS: Overall mean ± standard deviation (normalised to a median Hb of 13.8 mg/dl) was 57 ±5% for rcStO2 and 0.39 ±0.05 for rcFtO2E. Comparing no-hrPDA with hrPDA infants, there were no significant differences in mean rcStO2 (58 ±5% vs. 54 ±5%; p = .102), but in mean rcFtO2E (0.38 ±0.05 vs. 0.43 ±0.05; p = .038). Echocardiographic parameter and Doppler indices did not correlate with cerebral oxygenation. CONCLUSION: Oxygen transport capacity of the blood may confound NIRS data interpretation. Cerebral oxygenation determined by fd-NIRS provided additional information for PDA treatment decisions not offered by routine investigations. Whether indicating PDA therapy based on echocardiography complemented by data on cerebral oxygenation results in better outcomes should be investigated in future studies.


Assuntos
Encéfalo/diagnóstico por imagem , Permeabilidade do Canal Arterial/diagnóstico por imagem , Doenças do Prematuro/diagnóstico por imagem , Oxigênio/metabolismo , Espectroscopia de Luz Próxima ao Infravermelho , Biomarcadores/metabolismo , Encéfalo/metabolismo , Permeabilidade do Canal Arterial/metabolismo , Permeabilidade do Canal Arterial/fisiopatologia , Ecocardiografia Doppler , Feminino , Hemodinâmica , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/metabolismo , Doenças do Prematuro/fisiopatologia , Masculino , Estudos Prospectivos
9.
Rev Chil Pediatr ; 89(1): 10-17, 2018 Feb.
Artigo em Espanhol | MEDLINE | ID: mdl-29664498

RESUMO

New nutritional approaches to treat extreme premature babies have demonstrated relevant eviden ce of metabolic disturbances with early hypophosphatemia, especially in patients with intrauterine growth restriction (IUGR). They have shown late hypophosphatemia, as well, which is characteristic in the metabolic bone disease. A sytematic search of literature describing metabolic disturbances of phosphorus in preterm newborns is presented, related to the use of early parenteral nutrition and also in the context of metabolic bone disease. The articles were gathered from electronic data bases, such as PubMed and Rima. We include articles in english and spanish which were selected by titles and abstracts. Several strategies for early nutrition have been proposed in order to ensure an adequate amount of nutrients to accomplish the development and growth of preterm babies. Patients with parenteral nutrition support with different doses of phosphate, or inadequate calcium phosphate relation, or an increased amino acid content, may present hypophosphatemia, hypercalcemia, hy pomagnesemia, hypokalemia and hyperglycemia, all of these are additionally noteworthy in the pre sence of intrauterine growth restriction. Furthermore, said alterations are associated with prolonged mechanical ventilation, as well as bronchopulmonary dysplasia and increase in late onset sepsis. The late hypophosphatemia, described several years ago, arises as normocalcemia and as an increment of alkaline phosphatases in the metabolic bone disease in preterm babies, and also with an inadequate mineralization in different grades, secondary to an inadequate supply due to high nutritional requi rements in these patients. When early or late hypophosphatemia appears in preterm babies, it shall require timely control of phosphemia and will need to adjust the nutritional intake in order to correct it. In case of preterm babies with early parenteral nutrition it will also need a control of calcemia in the first week of birth, especially if those belonging to the IUGR group. Adjustment must be made along with metabolic follow up, as well. In late hypophosphatemia, a weekly or every two weeks fo llow up will be a must for all preterm babies in risk and they should be given supplements to get an optimum mineral supply.


Assuntos
Hipofosfatemia , Doenças do Prematuro , Biomarcadores/metabolismo , Cálcio/metabolismo , Retardo do Crescimento Fetal/fisiopatologia , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/etiologia , Hipofosfatemia/metabolismo , Hipofosfatemia/terapia , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/diagnóstico , Doenças do Prematuro/etiologia , Doenças do Prematuro/metabolismo , Doenças do Prematuro/terapia , Nutrição Parenteral/efeitos adversos , Fósforo/metabolismo , Síndrome da Realimentação/fisiopatologia
10.
Zhongguo Dang Dai Er Ke Za Zhi ; 20(7): 598-602, 2018 Jul.
Artigo em Zh | MEDLINE | ID: mdl-30022766

RESUMO

With the increase in the rescue success rate of critically ill preterm infants and extremely preterm infants, the incidence rate of bronchopulmonary dysplasia (BPD) is increasing year by year. BPD has a high mortality rate and high possibility of sequelae, which greatly affects the quality of life of preterm infants and brings a heavy burden to their families, and so the treatment of BPD is of vital importance. At present, no consensus has been reached on the treatment measures for BPD. However, recent studies have shown that early application of caffeine can prevent BPD. With reference to the latest studies on the effect of caffeine in the prevention of BPD, this article reviews the mechanism of action of caffeine in reducing pulmonary inflammation, improving morphological abnormalities of lung injury, reducing oxidative stress injury, and improving pulmonary function.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Cafeína/administração & dosagem , Doenças do Prematuro/prevenção & controle , Animais , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/fisiopatologia , Humanos , Recém-Nascido Prematuro/crescimento & desenvolvimento , Recém-Nascido Prematuro/metabolismo , Doenças do Prematuro/genética , Doenças do Prematuro/metabolismo , Doenças do Prematuro/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos
11.
J Pediatr ; 186: 49-56.e1, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28279433

RESUMO

OBJECTIVE: To characterize actual achieved patterns of oxygenation in infants born appropriate vs small for gestational age (SGA) randomized to a lower (85-89%) vs higher (91%-95%) oxygen saturation target in the Surfactant Positive Pressure and Oxygen Trial. To determine the association between achieved oxygen saturation levels and survival in infants born appropriate vs SGA enrolled in the Surfactant Positive Pressure and Oxygen Trial. STUDY DESIGN: Median oxygen saturation and intermittent hypoxemia events (<80%, 20 seconds-5 minutes) were documented in 1054 infants of 240/7-276/7 weeks of gestation while receiving supplemental oxygen during the first 3 days of life. RESULTS: Lower target infants who were small for gestational age had the lowest oxygen saturation and highest incidence of intermittent hypoxemia during the first 3 days of life. The lowest quartile of oxygen saturation (≤92%) during the first 3 days of life was associated with lower 90-day survival for both infants born appropriate and SGA. An increased incidence of intermittent hypoxemia events during the first 3 days of life was associated with lower 90-day survival only in infants born SGA. CONCLUSION: Lower achieved oxygen saturation during the first 3 days of life was associated with lower 90-day survival in extremely preterm infants. Infants born SGA had enhanced vulnerability to lower oxygen saturation targets as evidenced by lower achieved oxygen saturation and an association between increased intermittent hypoxemia events and lower survival. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00233324.


Assuntos
Pressão Positiva Contínua nas Vias Aéreas , Hipóxia/terapia , Doenças do Prematuro/metabolismo , Doenças do Prematuro/terapia , Oxigenoterapia , Surfactantes Pulmonares/uso terapêutico , Estudos de Coortes , Feminino , Humanos , Hipóxia/metabolismo , Hipóxia/mortalidade , Lactente Extremamente Prematuro , Recém-Nascido , Doenças do Prematuro/mortalidade , Recém-Nascido Pequeno para a Idade Gestacional , Masculino , Taxa de Sobrevida
12.
J Pediatr Gastroenterol Nutr ; 64(4): 550-554, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27467111

RESUMO

Preterm infants are at risk of developing gastrointestinal complications such as feeding intolerance and necrotizing enterocolitis. Near-infrared spectroscopy (NIRS) provides continuous monitoring of abdominal oxygenation (ArSO2) and could help to predict gastrointestinal complications in preterm neonates. In this prospective observational study, ArSO2 patterns at first enteral feed were evaluated by NIRS in 61 clinically stable preterm infants. Splanchnic-cerebral oxygenation ratio, which is a marker of gut hypoxia, was also calculated. ArSO2 and splanchnic-cerebral oxygenation ratio were significantly lower both at baseline and after feeding administration in infants who later developed feeding intolerance (n = 23). NIRS could help the early prediction of gastrointestinal complications in high-risk preterm infants.


Assuntos
Nutrição Enteral/efeitos adversos , Gastroenteropatias/etiologia , Doenças do Prematuro/etiologia , Oxigênio/metabolismo , Circulação Esplâncnica , Biomarcadores/metabolismo , Feminino , Gastroenteropatias/diagnóstico , Gastroenteropatias/metabolismo , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/diagnóstico , Doenças do Prematuro/metabolismo , Masculino , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Espectroscopia de Luz Próxima ao Infravermelho
13.
J Pediatr Gastroenterol Nutr ; 65(3): e47-e52, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27846067

RESUMO

OBJECTIVES: Several studies associated altered gut microbiota composition in preterm infants with late-onset sepsis (LOS), up to days before clinical onset of sepsis. Microbiota analysis as early diagnostic biomarker is, however, in clinical practice currently not feasible because of logistic aspects and high costs. Therefore, we hypothesized that analysis of fecal volatile organic compounds (VOCs) may serve as noninvasive biomarker to predict LOS at a preclinical stage, because VOC reflect the composition and activity of intestinal microbial communities. METHODS: In a prospective multicenter study, fecal samples were collected daily from infants with a gestational age of <30 weeks. VOC signatures of fecal samples from infants with LOS, collected up to 5 days before diagnosis, were analyzed by means of an electronic nose technology (Cyranose 320) and compared to matched controls. RESULTS: Fecal VOC profiles of infants with LOS (n = 36) could be discriminated from controls (n = 40) at 3 days (area under the curve [±95% confidence interval], P value, sensitivity, specificity; 70.2 [52.2-88.3], 0.033, 57.1%, 61.5%), 2 days (77.7 [62.7-92.7], 0.050, 75.0%, 70.8%), and 1 day (70.4 [49.6-91.3], 0.037, 64.3%, 64.3%) before the onset of LOS. CONCLUSIONS: Fecal VOC profiles of preterm infants with LOS could be discriminated from matched controls, up to 3 days before clinical onset of the disease, underlining the hypothesis that intestinal microbiota may play an etiological role in LOS. Notably, VOC profiling is clinically feasible and the potential of this technique in the early detection of LOS needs to be confirmed in future studies.


Assuntos
Fezes/química , Microbioma Gastrointestinal , Doenças do Prematuro/diagnóstico , Sepse Neonatal/diagnóstico , Compostos Orgânicos Voláteis/metabolismo , Biomarcadores/metabolismo , Estudos de Casos e Controles , Nariz Eletrônico , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/metabolismo , Doenças do Prematuro/microbiologia , Masculino , Sepse Neonatal/metabolismo , Sepse Neonatal/microbiologia , Estudo de Prova de Conceito , Estudos Prospectivos
14.
Rev Chil Pediatr ; 88(4): 487-494, 2017.
Artigo em Espanhol | MEDLINE | ID: mdl-28898316

RESUMO

BACKGROUND: Metabolic bone disease (MBD) of prematurity is a complication of multifactorial aetiology, which has been increasing, due to progressive decrease in mortality of preterm newborns. The aim of the study was to analyze risk factors of severe MBD and its analytical markers. PATIENTS AND METHOD: Retrospective study involving preterm infants less than 32 weeks gestational age and/or weight less tan 1,500 g born between january 2012 and december 2014. Comparison was made according to the presence of severe MBD. RESULTS: 139 patients were recruited. Mean value of 25(OH)D3 was 70.68 ± 25.20 nmol/L, being higher in patients born in spring-summer than in autumn-winter (80.94 ± 25.33 vs 61.13 ± 21.07; p = 0.000). Levels of 25(OH)D3 were similar in patients with severe MBD compared with the rest of patients (65.61 ± 26.49 vs 72.07 ± 24.89, P = 0.283). Higher levels of alkaline phosphatase (AP, IU/L ) (1314.19 ± 506.67 vs 476.56 ± 188.85; p = 0.000) were found in these patients. Cutoff point of AP 796.5 IU/L (S 95.2%, specificity 92.4%) was calculated by ROC curve. The risk factors most associated to severe EMO were restricted fetal growth, birth weight, duration of ventilation therapy and parenteral nutrition. CONCLUSIONS: AP levels were the best marker of severe MBD development. EMO risk increases with the number of risk factors and lower levels of 25(OH)D3. Levels of 25(OH)D3 higher than 70nmol/L appear to protect from the development of severe MBD, even in patients with multiple risk factors.


Assuntos
Doenças Ósseas Metabólicas/diagnóstico , Doenças Ósseas Metabólicas/etiologia , Doenças do Prematuro/diagnóstico , Doenças do Prematuro/etiologia , Biomarcadores/metabolismo , Doenças Ósseas Metabólicas/metabolismo , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/metabolismo , Masculino , Estudos Retrospectivos , Fatores de Risco
15.
Transfusion ; 56(4): 808-15, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26643925

RESUMO

BACKGROUND: Extremely preterm infants receive frequent blood transfusions in the first week of life. The aim of this study was to measure the effect of blood transfusion on intestinal blood flow and oxygenation during the first week of life in extremely preterm infants. STUDY DESIGN AND METHODS: Superior mesenteric artery (SMA) peak systolic velocity (PSV) and diastolic velocities were measured 30 to 60 minutes before and after transfusion. Splanchnic tissue hemoglobin index (sTHI), splanchnic tissue oxygenation index (sTOI), and splanchnic fractional tissue oxygen extraction (sFTOE) were measured continuously from 15 to 20 minutes before to after transfusion along with vital variables. RESULTS: Twenty infants were studied (median gestational age, 26 weeks). Ten infants were partially fed (15-68 mL/kg/day). Heart rate and SaO2 remained unaltered; blood pressure increased significantly (p < 0.01) after transfusion. Mean SMA PSV (p = 0.63) and diastolic velocity (p = 0.65) remained unaltered. Mean pretransfusion SMA PSV was similar in partially fed (0.78 m/sec) compared to unfed infants (0.52 m/sec; p = 0.06) and the response to transfusion was not dissimilar. There was a significant increase in sTHI (mean difference, 32.3%; p < 0.01) and sTOI (14.6%; p = 0.03) and decrease in sFTOE (22.1%; p < 0.01) after transfusion. There was no significant difference in sTHI or sTOI between fed and unfed infants and their response to transfusion. CONCLUSIONS: Blood transfusion increased blood pressure and intestinal tissue oxygenation but did not alter blood flow velocities. Partial feeding had no impact on intestinal blood flow and tissue oxygenation changes.


Assuntos
Transfusão de Sangue , Lactente Extremamente Prematuro , Mucosa Intestinal/metabolismo , Intestinos/irrigação sanguínea , Consumo de Oxigênio , Fluxo Sanguíneo Regional/fisiologia , Circulação Esplâncnica/fisiologia , Velocidade do Fluxo Sanguíneo/fisiologia , Pressão Sanguínea , Feminino , Idade Gestacional , Frequência Cardíaca , Humanos , Lactente Extremamente Prematuro/fisiologia , Recém-Nascido , Doenças do Prematuro/diagnóstico por imagem , Doenças do Prematuro/metabolismo , Doenças do Prematuro/fisiopatologia , Doenças do Prematuro/terapia , Intestinos/diagnóstico por imagem , Masculino , Ultrassonografia
16.
Childs Nerv Syst ; 32(11): 2097-2103, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27541865

RESUMO

Proinflammatory cytokines are essential mediators and indicators of an inflammatory process occurring in the body. Their physiological role is to stimulate the immune response, yet their excessive propagation and interaction with cells outside the immune system may be linked to the risk of organ damage. This is specifically important in the case of immature tissues of fetuses and prematurely born infants. Analysis of the concentrations of specific cytokines in different compartments makes it possible to assess the risk of premature birth, preterm rupture of the membranes, and to determine an existing intrauterine infection. The purpose of this paper is to summarize the existing research concerning the relationships between the concentrations of specific proinflammatory cytokines in different compartments (maternal blood serum, amniotic fluid, umbilical cord blood, arterial and venous blood, and cerebrospinal fluid of the newborn) and the risk of intraventricular hemorrhage (IVH) and the degree of its severity. The paper takes also into account the assessment of the usefulness of cytokines as biomarkers for IVH and its complications (posthemorrhagic hydrocephalus, white matter injury).


Assuntos
Hemorragia Cerebral/etiologia , Hemorragia Cerebral/metabolismo , Citocinas/metabolismo , Doenças do Prematuro/etiologia , Doenças do Prematuro/metabolismo , Recém-Nascido Prematuro , Humanos , Recém-Nascido
17.
Am J Perinatol ; 33(2): 130-5, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26344008

RESUMO

BACKGROUND: Haptoglobin (Hp) is an acute phase protein with antioxidant, bacteriostatic, and anti-inflammatory activities. Hp proteins associated with the three major phenotypes differ in their proinflammatory and anti-inflammatory action. Inflammation and oxidative stress are both involved in most pathophysiological processes in premature infants. The objective of this study was to determine whether Hp phenotype influences clinical manifestations and sepsis incidence in the premature infants. OBJECTIVE: Infants born before 35 weeks gestational age were prospectively evaluated for Hp phenotype and clinical events, including sepsis, necrotizing enterocolitis, bronchopulmonary dysplasia, intraventricular hemorrhage, and retinopathy of prematurity. The participants were observed until discharge. METHODS: A total of 122 preterm infants were enrolled in the study. Clinical events were not affected by the Hp phenotype. The expression of Hp protein was extremely low in the study population. More septic episodes were found in infants with a birth weight greater than 1,500 g, although, the difference was not statistically significant. RESULTS: Extremely low expression of Hp may explain the lack of a correlation between Hp phenotype and sepsis in preterm infants. Further research involving a larger neonatal population is required to better understand the role of the Hp phenotype in morbidity of premature infants.


Assuntos
Displasia Broncopulmonar/epidemiologia , Hemorragia Cerebral/epidemiologia , Enterocolite Necrosante/epidemiologia , Haptoglobinas/metabolismo , Retinopatia da Prematuridade/epidemiologia , Sepse/epidemiologia , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Estudos de Coortes , Eletroforese em Gel de Poliacrilamida , Enterocolite Necrosante/genética , Enterocolite Necrosante/metabolismo , Feminino , Haptoglobinas/genética , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/epidemiologia , Doenças do Prematuro/genética , Doenças do Prematuro/metabolismo , Israel/epidemiologia , Masculino , Fenótipo , Reação em Cadeia da Polimerase , Estudos Prospectivos , Fatores de Proteção , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/metabolismo , Fatores de Risco , Sepse/genética , Sepse/metabolismo
18.
Biochim Biophys Acta ; 1841(3): 362-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24120574

RESUMO

Fatty acids in the epidermis can be incorporated into complex lipids or exist in a free form, and they are crucial to proper functions of the epidermis and its appendages, such as sebaceous glands. Epidermal fatty acids can be synthesized de novo by keratinocytes or taken up from extracutaneous sources in a process that likely involves protein transporters. Several proteins that are expressed in the epidermis have been proposed to facilitate the uptake of long-chain fatty acids (LCFA) in mammalian cells, including fatty acid translocase/CD36, fatty acid binding protein, and fatty acid transport protein (FATP)/very long-chain acyl-CoA synthetase. In this review, we will discuss the mechanisms by which these candidate transporters facilitate the uptake of fatty acids. We will then discuss the clinical implications of defects in these transporters and relevant animal models, including the FATP4 animal models and ichthyosis prematurity syndrome, a congenital ichthyosis caused by FATP4 deficiency. This article is part of a Special Issue entitled The Important Role of Lipids in the Epidermis and their Role in the Formation and Maintenance of the Cutaneous Barrier. Guest Editors: Kenneth R. Feingold and Peter Elias.


Assuntos
Aniridia , Proteínas de Transporte de Ácido Graxo , Ácidos Graxos , Ictiose , Doenças do Prematuro , Rim/anormalidades , Transtornos Psicomotores , Pele , Animais , Aniridia/genética , Aniridia/metabolismo , Aniridia/patologia , Transporte Biológico Ativo/genética , Modelos Animais de Doenças , Proteínas de Transporte de Ácido Graxo/genética , Proteínas de Transporte de Ácido Graxo/metabolismo , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Feminino , Humanos , Ictiose/genética , Ictiose/metabolismo , Ictiose/patologia , Doenças do Prematuro/genética , Doenças do Prematuro/metabolismo , Doenças do Prematuro/patologia , Rim/metabolismo , Rim/patologia , Masculino , Transtornos Psicomotores/genética , Transtornos Psicomotores/metabolismo , Transtornos Psicomotores/patologia , Pele/metabolismo , Pele/patologia
19.
Neurochem Res ; 40(12): 2527-43, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26148530

RESUMO

Brain development is a highly orchestrated complex process. The developing brain utilizes many substrates including glucose, ketone bodies, lactate, fatty acids and amino acids for energy, cell division and the biosynthesis of nucleotides, proteins and lipids. Metabolism is crucial to provide energy for all cellular processes required for brain development and function including ATP formation, synaptogenesis, synthesis, release and uptake of neurotransmitters, maintaining ionic gradients and redox status, and myelination. The rapidly growing population of infants and children with neurodevelopmental and cognitive impairments and life-long disability resulting from developmental brain injury is a significant public health concern. Brain injury in infants and children can have devastating effects because the injury is superimposed on the high metabolic demands of the developing brain. Acute injury in the pediatric brain can derail, halt or lead to dysregulation of the complex and highly regulated normal developmental processes. This paper provides a brief review of metabolism in developing brain and alterations found clinically and in animal models of developmental brain injury. The metabolic changes observed in three major categories of injury that can result in life-long cognitive and neurological disabilities, including neonatal hypoxia-ischemia, pediatric traumatic brain injury, and brain injury secondary to prematurity are reviewed.


Assuntos
Lesões Encefálicas/metabolismo , Encéfalo/crescimento & desenvolvimento , Adulto , Animais , Química Encefálica , Lesões Encefálicas/complicações , Lesões Encefálicas/congênito , Metabolismo Energético , Feminino , Humanos , Doenças do Prematuro/metabolismo , Gravidez
20.
Acta Paediatr ; 104(7): 657-62, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25382627

RESUMO

AIM: Even short periods of hyperoxia may induce prolonged cerebral vasoconstriction in newborn infants, and this could theoretically lead to cerebral ischaemia even once normoxia is re-established. This study aimed to investigate the effect of brief hyperoxic exposures on regional cerebral tissue oxygen saturation (rStO2 ) and to evaluate whether any observed prolonged cerebral vasoconstriction was related to maturity. METHODS: The study included 30 infants with a postmenstrual age of more than 32 weeks, who were treated with nasal continuous positive airway pressure and a fraction of inspired oxygen of ≤0.3. The INVOS 5100C oximeter was used to measure rStO2 before, during and after two hyperoxic exposures. If hyperoxia induced a prolonged cerebral vasoconstriction, posthyperoxic rStO2 would be expected to decrease. RESULTS: rStO2 increased slightly after the first hyperoxic exposure, with a mean difference of 1.37% (95% CI 0.15, 2.6). After the second oxygen exposure, rStO2 remained unchanged with a mean difference of -0.4% (95% CI -1.6, 0.78). Differences in rStO2 were not related to gestational age in either of the two hyperoxic episodes. CONCLUSION: We found no evidence to support the theory that transient hyperoxia induces prolonged cerebral vasoconstriction in infants with a postmenstrual age above 32 weeks.


Assuntos
Circulação Cerebrovascular/fisiologia , Hiperóxia/metabolismo , Hiperóxia/terapia , Doenças do Prematuro/metabolismo , Doenças do Prematuro/terapia , Oxigenoterapia , Fatores Etários , Pressão Positiva Contínua nas Vias Aéreas , Feminino , Humanos , Hiperóxia/fisiopatologia , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/fisiopatologia , Masculino , Oximetria , Consumo de Oxigênio/fisiologia , Vasoconstrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa