Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Cell Mol Med ; 24(10): 5565-5577, 2020 05.
Article in English | MEDLINE | ID: mdl-32301278

ABSTRACT

Malignant mesothelioma (MM) is an aggressive asbestos-related cancer of the serous membranes. Despite intensive treatment regimens, MM is still a fatal disease, mainly due to the intrinsic resistance to current therapies and the lack of predictive markers and new valuable molecular targets. Protein arginine methyltransferase 5 (PRMT5) inhibition has recently emerged as a potential therapy against methylthioadenosine phosphorylase (MTAP)-deficient cancers, in which the accumulation of the substrate 5'-methylthioadenosine (MTA) inhibits PRMT5 activity, thus sensitizing the cells to further PRMT5 inhibition. Considering that the MTAP gene is frequently codeleted with the adjacent cyclin-dependent kinase inhibitor 2A (CDKN2A) locus in MM, we assessed whether PRMT5 could represent a therapeutic target also for this cancer type. We evaluated PRMT5 expression, the MTAP status and MTA content in normal mesothelial and MM cell lines. We found that both administration of exogenous MTA and stable PRMT5 knock-down, by short hairpin RNAs (shRNAs), selectively reduced the growth of MTAP-deleted MM cells. We also observed that PRMT5 knock-down in MTAP-deficient MM cells reduced the expression of E2F1 target genes involved in cell cycle progression and of factors implicated in epithelial-to-mesenchymal transition. Therefore, PRMT5 targeting could represent a promising new therapeutic strategy against MTAP-deleted MMs.


Subject(s)
Gene Deletion , Gene Expression Regulation, Neoplastic , Gene Silencing , Mesothelioma/genetics , Protein-Arginine N-Methyltransferases/genetics , Purine-Nucleoside Phosphorylase/genetics , Cell Line, Tumor , Chromatography, Liquid , Epithelial-Mesenchymal Transition/genetics , Gene Knockdown Techniques , Humans , Immunohistochemistry , Mesothelioma/metabolism , Mesothelioma/pathology , Tandem Mass Spectrometry
2.
Curr Med Res Opin ; 35(sup1): 17-20, 2019 03.
Article in English | MEDLINE | ID: mdl-30864896

ABSTRACT

INTRODUCTION: We investigated the effectiveness of sacubitril/valsartan by performing laboratory tests and a 6-minute walking test (6-MWT) at 1 and 6 months after treatment initiation. METHODS: We evaluated patients admitted to our Cardiology Department, stabilized after an episode of acute decompensated heart failure (HF), who were considered eligible for sacubitril/valsartan therapy. Therapy was initiated after interrupting angiotensin-converting enzyme (ACE) inhibitors for at least 36 h or after the last dose of an angiotensin receptor blocker (ARB). In naïve patients, we initiated a low dose of sacubitril/valsartan combination following patient stabilization. Before discharge, a 6-MWT was performed to evaluate patient's functional capacity, measuring total walked distance (in meters), oxygen saturation and heart rate at the beginning and at the end of the test; Borg Scale was applied to evaluate the intensity of dyspnoea. After discharge, follow-up visits at 1 and 6 months, 2D-echocardiography, blood tests and 6-MWT were performed to re-evaluate the efficacy of the treatment. RESULTS: A total of 14 patients (85.7% males) were included. Mean age was 66.0 ± 10.3 years. Body mass index (BMI) was 29.9 ± 4.7 kg/m2. There were no differences in creatinine at admission compared with values at 1 and 6 months. Mean left ventricular ejection fraction (LVEF) was 28.7 ± 4.7% at baseline and increased to 33.5 ± 6.6% and 38.0 ± 2.9% at 1 and 6 months, respectively (p = .028). Total distance covered at 6-MWT increased over the study period (baseline: 227.4 ± 62.8 m; 6 months: 257.3 ± 65.2 m, p = .317) although the increase was not statistically significant. CONCLUSIONS: The present experience showed that angiotensin receptor-neprilysin inhibitor (ARNi) might represent a new valuable therapeutic strategy, even at the earlier stages of stabilized acute HF. Therefore, we suggest a clinical practice algorithm, to consider before discharge, which should be validated by further analyses.


Subject(s)
Aminobutyrates/therapeutic use , Angiotensin Receptor Antagonists/therapeutic use , Heart Failure/drug therapy , Stroke Volume/physiology , Tetrazoles/therapeutic use , Aged , Biphenyl Compounds , Drug Combinations , Female , Heart Failure/physiopathology , Humans , Male , Middle Aged , Valsartan
3.
J Perinat Med ; 47(1): 82-89, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30110254

ABSTRACT

Background Oxidative stress plays an important part in the pathophysiology of hypoxic-ischemic encephalopathy (HIE) and is reliably measured through prostanoids following lipid peroxidation of polyunsaturated fatty acids (PUFAs). The aim of the study is to measure oxidative stress in the prefrontal cortex, white matter and hippocampus in the brains of hypoxic-ischemic piglets treated with docosahexaenoic acid (DHA) and therapeutic hypothermia (TH) and investigate the additive effects of DHA on hypothermia by factorial design. Methods Fifty-five piglets were randomized as having severe global hypoxia (n=48) or not (sham, n=7). Hypoxic piglets were further randomized: vehicle (VEH), DHA, VEH+hypothermia (HT) or HT+DHA. A total of 5 mg/kg DHA was given intravenously 210 min after the end of hypoxia. Brain tissues were analyzed using liquid chromatography triple quadrupole mass spectrometry technique (LC-MS). A two-way analysis of variance (ANOVA) was performed with DHA and HT as main effects. Results In the white matter, we found main effects of DHA on DH-isoprostanes (P=0.030) and a main effect of HT on F4-neuroprostanes (F4-NeuroPs) (P=0.007), F2-isoprostanes (F2-IsoPs) (P=0.043) and DH-isoprostanes (P=0.023). In the cortex, the ANOVA analysis showed the interactions of main effects between DHA and HT for neurofuranes (NeuroFs) (P=0.092) and DH-isoprostanes (P=0.015) as DHA significantly reduced lipid peroxidation in the absence of HT. DHA compared to VEH significantly reduced NeuroFs (P=0.019) and DH-isoprostanes (P=0.010). No differences were found in the hippocampus. Conclusion After severe hypoxia, HT reduced lipid peroxidation in the white matter but not in the cortical gray matter. HT attenuated the reducing effect of DHA on lipid peroxidation in the cortex. Further studies are needed to determine whether DHA can be an effective add-on therapy for TH.


Subject(s)
Fatty Acids, Unsaturated , Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain , Lipid Peroxidation , Oxidative Stress , Animals , Animals, Newborn , Chromatography, Liquid/methods , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Female , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/prevention & control , Isoprostanes/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Mass Spectrometry/methods , Oxidative Stress/drug effects , Oxidative Stress/physiology , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Pregnancy , Swine , Treatment Outcome , White Matter/diagnostic imaging , White Matter/metabolism
4.
Oxid Med Cell Longev ; 2018: 7483062, 2018.
Article in English | MEDLINE | ID: mdl-30140369

ABSTRACT

During the perinatal period, free radicals (FRs) are involved in several physiological roles such as the cellular responses to noxia, the defense against infectious agents, the regulation of cellular signaling function, and the induction of a mitogenic response. However, the overproduction of FRs and the insufficiency of an antioxidant mechanism result in oxidative stress (OS) which represents a deleterious process and an important mediator of damage to the placenta and the developing fetus. After birth, OS can be magnified by other predisposing conditions such as hypoxia, hyperoxia, ischemia, hypoxia ischemia-reperfusion, inflammation, and high levels of nonprotein-bound iron. Newborns are particularly susceptible to OS and oxidative damage due to the increased generation of FRs and the lack of adequate antioxidant protection. This impairment of the oxidative balance has been thought to be the common factor of the so-called "free radical related diseases of prematurity," including retinopathy of prematurity, bronchopulmonary dysplasia, intraventricular hemorrhage, periventricular leukomalacia, necrotizing enterocolitis, kidney damage, and oxidative hemolysis. In this review, we provide an update focused on the factors influencing these diseases refining the knowledge about the role of OS in their pathogenesis and the current evidences of such relationship. Mechanisms governing FR formation and subsequent OS may represent targets for counteracting tissue damage.


Subject(s)
Free Radicals/adverse effects , Infant, Premature, Diseases/etiology , Humans , Infant, Premature, Diseases/pathology , Oxidative Stress
5.
Cytokine ; 111: 119-124, 2018 11.
Article in English | MEDLINE | ID: mdl-30142532

ABSTRACT

PURPOSE: Inflammation is a crucial but understudied mechanism of neuronal injury after hypoxia-ischemia. The aim was to identify a panel of cytokines involved in brain injury in neonates with hypoxic ischemic encephalopathy (HIE). METHODS: Ten newborns with HIE undergoing to therapeutic hypothermia (TH, HIE Group) and 8 healthy newborns (CTRL Group) were enrolled. For the HIE group, 5 samples were collected: between 0 and 6 h of life (time 1), 12 h (time 2), 24 h (time 3), 48 h (time 4) and 96 h of life (time 5). For the CTRL group, one sample was collected. A panel of 48 inflammatory cytokines was determined in all samples. Data were analyzed using multivariate statistical analysis (Principal component analysis, PCA) RESULTS: 17 cytokines, among 48 analyzed, were found to be significantly different, initially, between the CTRL and HIE groups: 12 with reported pro-inflammatory effects and 5 with reported anti-inflammatory effects. In the HIE group cytokines showed a decreasing trend during the TH and at the end of treatment comparable to the CTRL group. IL-18 did demonstrate a slight increase at time 3 during HT but decreased steadily at sampling times, 4 and 5. CONCLUSIONS: Our data demonstrates that many pathways of the inflammatory cascade are activated following hypoxic-ischemic injury. This information will increase our understanding of changes in cytokines over time in neonates with HIE undergoing TH.


Subject(s)
Cytokines , Hypothermia, Induced , Hypoxia-Ischemia, Brain , Cytokines/biosynthesis , Cytokines/immunology , Female , Humans , Hypoxia-Ischemia, Brain/blood , Hypoxia-Ischemia, Brain/immunology , Hypoxia-Ischemia, Brain/therapy , Infant, Newborn , Male , Retrospective Studies
6.
Oxid Med Cell Longev ; 2018: 7608108, 2018.
Article in English | MEDLINE | ID: mdl-30050660

ABSTRACT

This research paper is aimed at evaluating the predictive role of a default panel of oxidative stress (OS) biomarkers for the early identification of infants at high risk of HIE and their validation through the correlation with MRI findings. A multicenter prospective observational study was performed between March 2012 and April 2015 in two European tertiary NICUs. Eighty-four term infants at risk for HIE (pH < 7, BE < -13 mmol/L, and 5' Apgar < 5) were enrolled. Three were excluded for chromosomal abnormalities and one due to lack of blood samples. The final population was divided according to the severity of perinatal hypoxia into 2 groups: mild/moderate HIE and severe HIE. Advanced oxidation protein products (AOPP), non-protein-bound iron (NPBI), and F2-isoprostanes (F2-IsoPs) were measured in blood samples at P1 (4-6 hours), P2 (24-72 hours), and P3 (5 days), in both groups. MRIs were scored for the severity of brain injury, using a modified Barkovich score. The mean GA was 39.8 weeks (SD 1.4) and the mean birth weight 3538 grams (SD 660); 37 were females and 43 males. Significantly lower 5' Apgar score, pH, and BE and higher Thompson score were found in group II compared to group I at birth. Group II showed significantly higher AOPP and NPBI levels than group I (mean (SD) AOPP: 15.7 (15.5) versus 34.1 (39.2), p = 0.033; NPBI 1.1 (2.5) versus 3.9 (4.4), p = 0.013) soon after birth (P1). No differences were observed in OS biomarker levels between the two groups at P2 and P3. A regression model, including adjustment for hypothermia treatment, gender, and time after birth, showed that AOPP levels and male gender were both risk factors for higher brain damage scores (AOPP: OR 3.6, 95% CI (1.1-12.2) and gender: OR 5.6, 95% CI (1.2-25.7), resp.). Newborns with severe asphyxia showed higher OS than those with mild asphyxia at birth. AOPP are significantly associated with the severity of brain injury assessed by MRI, especially in males.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Oxidative Stress/physiology , Advanced Oxidation Protein Products/blood , Biomarkers/metabolism , Brain Injuries/blood , Brain Injuries/metabolism , F2-Isoprostanes/blood , Female , Humans , Hypoxia-Ischemia, Brain/blood , Infant, Newborn , Magnetic Resonance Imaging , Male , Prospective Studies
7.
Mediators Inflamm ; 2018: 2845352, 2018.
Article in English | MEDLINE | ID: mdl-29706798

ABSTRACT

BACKGROUND: Rooming-in practice improves breastfeeding and reduces newborn stress reactivity. When this modality is not available, partial rooming-in after birth can be considered. Salivary cortisol levels (SCLs) are considered reliable biomarkers to indicate stress. OBJECTIVE: To test the hypothesis that rooming-in duration impacts neonatal stress response in hospitalized newborns. DESIGN/METHODS: Forty term newborns, enrolled in the Neonatology and Obstetrics Nursing, C.G. Ruesch, Naples, Italy, were divided, according to the mother's choice, into the study (SG; n = 20) and control (CG; n = 20) groups if they received full (24 hs) or partial (14 hs) rooming-in care, respectively. Saliva samples were collected from all babies between 7:00 a.m. and 8:00 a.m. of the 3rd day of life by using oral swab. Salivary cortisol levels were measured using an enzyme immunoassay kit (Salimetrics LLC, PA, USA). RESULTS: A statistically significant difference in the SCLs between SG and CG was found (median: 258 ng/dl versus 488.5 ng/dl; p = 0.048). CONCLUSIONS: Data support the practice of full rooming-in care compared with partial rooming-in. The rooming-in duration clearly reduces SCLs and likely neonatal stress. These lower SCLs may have long-term positive effects reducing the risk of metabolic syndrome, high blood pressure, and cognitive and behavioural changes.


Subject(s)
Hydrocortisone/analysis , Rooming-in Care/methods , Saliva/chemistry , Adult , Breast Feeding , Female , Humans , Infant, Newborn , Male , Pregnancy
8.
Molecules ; 22(12)2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29194416

ABSTRACT

Melatonin possesses potential efficacy in perinatal brain injuries, and has been proposed as adjunctive pharmacological therapy in combination with hypothermia in the clinical setting. However, the pharmacokinetics of melatonin in preterm and term newborns is still unknown. The aim of this study was to analyze the pharmacokinetics of melatonin after intragastric administration in preterm infants. Preterm newborns were enrolled 24-72 h after birth, and randomly assigned to three groups receiving a single bolus of 0.5 mg·kg-1 melatonin, or 3 boluses of 1 or 5 mg·kg-1 of melatonin at 24-h intervals. Blood samples were collected before and at selective times after melatonin administration. The half-life of melatonin in plasma ranged from 7.98 to 10.94 h, and the area under the curve (AUC) from 10.48 to 118.17 µg·mL-1·h-1. Our results indicate a different pharmacokinetic profile in premature newborns, compared to adults and experimental animals. The high peak plasma concentrations and the long half-life indicate that in the neonatal clinical setting, it is possible to obtain and maintain high serum concentrations using a single administration of melatonin repeated every 12/24 h.


Subject(s)
Melatonin/pharmacokinetics , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Combined Modality Therapy , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Infant , Infant, Newborn , Infant, Premature , Melatonin/administration & dosage , Melatonin/blood , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/blood , Pregnancy
9.
Mediators Inflamm ; 2017: 1758432, 2017.
Article in English | MEDLINE | ID: mdl-28512386

ABSTRACT

Oxidative stress (OS) is a common pathogenic factor involved in the onset of several diseases in humans, from immunologic disorders to malignancy, being a serious public health problem. In perinatal period, OS has been associated with adverse outcome of pregnancy and neonatal diseases. Dangerous effects of OS are mediated by increased production of free radicals (FRs) following various mechanisms, such as hypoxia, ischemia reperfusion, hyperoxia, inflammation, mitochondrial dysfunction, Fenton chemistry, and prostaglandin metabolism. FRs have short half-life, and their measurement in vivo is faced with many challenges. However, oxyradical derivatives are stable and thus may be measured and monitored repeatedly. The quantification of OS is based on the measurement of specific biomarkers in biologic fluids and tissues, which reflect induced oxidative damage to lipids, proteins, and DNA. Prostanoids, non-protein-bound iron (NPBI), and advanced oxidation protein products (AOPP) are actually considered truly specific and reliable for neonatal injury. Defining reference values for these biomarkers is necessary to investigate their role in neonatal diseases or also to evaluate the success of treatments. In this work, we wanted to define laboratory reference values for biomarkers of OS in a healthy population of term newborns.


Subject(s)
Advanced Oxidation Protein Products/metabolism , Biomarkers/metabolism , Fetal Blood/metabolism , Isoprostanes/metabolism , Oxidative Stress/physiology , Adult , Advanced Oxidation Protein Products/standards , Female , Humans , Infant, Newborn , Isoprostanes/standards , Male , Tandem Mass Spectrometry
10.
Neonatology ; 112(1): 1-8, 2017.
Article in English | MEDLINE | ID: mdl-28142140

ABSTRACT

BACKGROUND: Perinatal hypoxic-ischemic brain damage is a major cause of acute mortality and chronic neurological morbidity in infants and children. Oxidative stress due to free radical formation and the initiation of abnormal oxidative reactions appears to play a key role. Docosahexanoic acid (DHA), a main component of brain membrane phospholipids, may act as a neuroprotectant after hypoxia-ischemia by regulating multiple molecular pathways and gene expression. OBJECTIVES: The aims of this study were to test the hypothesis that DHA provides significant protection against lipoperoxidation damage in the cerebral cortex and hippocampus in a neonatal piglet model of severe hypoxia-reoxygenation. METHODS: Newborn piglets, Noroc (LYLD), were subjected to severe global hypoxia. One group was resuscitated with ambient air (21% group, n = 11) and another also received 5 mg/kg of DHA 4 h after the end of hypoxia (21% DHA group, n = 10). After 9.5 h, tissues from the prefrontal cortex and hippocampus were sampled and the levels of isoprostanes, neuroprostanes, neurofurans, and F2-dihomo-isoprostanes were determined by the liquid chromatography triple quadrupole mass spectrometry technique. RESULTS: Lipid peroxidation biomarkers were significantly lower in both the cortex and hippocampus in the DHA-treated group compared with the untreated group. CONCLUSIONS: The present study demonstrates that DHA administration after severe hypoxia in newborn piglets has an antioxidative effect in the brain, suggesting a protective potential of DHA if given after injuries to the brain.


Subject(s)
Antioxidants/pharmacology , Asphyxia Neonatorum/drug therapy , Docosahexaenoic Acids/pharmacology , Hippocampus/drug effects , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Prefrontal Cortex/drug effects , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Biomarkers/metabolism , Disease Models, Animal , Furans/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Isoprostanes/metabolism , Lipid Peroxidation/drug effects , Neuroprostanes/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Sus scrofa
11.
Int J Angiol ; 25(5): e4-e7, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28031640

ABSTRACT

We report a clinical case of a 45-year-old male with a diagnosis of inferior myocardial infarction and previous history of rheumatic fever during his childhood. Coronary angiography demonstrated normal coronary arteries. Transthoracic echocardiogram showed hypokinetic left ventricular inferolateral wall and mitral stenosis; furthermore, speckle tracking analysis revealed reduction of global longitudinal strain involving the inferior wall. A three-dimensional transesophaegeal echocardiography, performed to better characterize the anatomy of the valve and to find possible source of embolic infarct in an enlarged left atrium, showed rheumatic valvular involvement. Cardiac magnetic resonance confirmed the ischemic damage and also provided prognostic information. A multimodality imaging approach should be mandatory in patients with acute myocardial infarction and normal coronary angiography, to define possible sources of embolic infarction and to quantify myocardial damage.

12.
Placenta ; 46: 72-78, 2016 10.
Article in English | MEDLINE | ID: mdl-27697224

ABSTRACT

BACKGROUND: Prenatal conditions of enhanced oxidative stress (OS) linked to inflammation or hypoxia have been associated with impaired fetal growth and preterm delivery. Little is known regarding biomarkers of OS in the cord blood of preterm infants and placental histological patterns. OBJECTIVES: To test the hypothesis that placental lesions indicating chorioamnionitis (CA) or vascular underperfusion (VU) are associated with increased OS in the offspring. METHODS: 120 neonates born below 29+6 weeks of gestational age (GA) were enrolled. Histological characteristics of placentas from their mothers were classified as normal (CTRL group), histological CA (HCA) and vascular underperfusion (VU). Serum concentrations of isoprostanes (IsoPs), non-protein bound iron (NPBI) and advanced oxidative protein products (AOPP), were determined in cord blood. RESULTS: IsoPs, NPBI and AOPP were significantly increased in HCA group compared to CTRL group. The multivariable regression model, adjusted for GA, maternal age, parity, maternal diabetes, maternal obesity and presence/absence of fetal growth restriction (FGR), showed a significant association between the presence of HCA and increased OS biomarkers levels in cord blood (IsoPs: p = 0.006; NPBI: p = 0.014; AOPP: p = 0.007). Placental VU lesions were significantly associated with higher umbilical IsoPs, NPBI and AOPP levels (IsoPs: p = 0.008; NPBI: p = 0.002; AOPP: p = 0.040). In the cases of placental VU lesions associations were also found between high AOPP levels and low GA (p = 0.002) and the presence of fetal growth restriction (p = 0.014). CONCLUSIONS: Placental lesions indicating inflammation or impaired perfusion are associated with higher cord blood levels of OS biomarkers explaining the fetal susceptibility to oxidative injury and the need of antioxidant protection.


Subject(s)
Chorioamnionitis/pathology , Infant, Premature/blood , Oxidative Stress , Placenta/pathology , Premature Birth/pathology , Adult , Female , Humans , Infant, Newborn , Placental Circulation , Pregnancy , Prospective Studies
13.
J Matern Fetal Neonatal Med ; 27 Suppl 2: 27-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25284174

ABSTRACT

OBJECTIVE: To investigate changes in global metabolic profile between: 1 - breast milk and formula milk, 2 - breast milk from mothers delivering at different gestational age (GA) collected within one week from delivery, and then week by week until term equivalent age. METHODS: Proton magnetic resonance spectroscopy (MRS) was used to analyze the water-soluble and lipid fractions extracted from 50 milk samples, 46 human milk at different GA, from 23 weeks of gestation until term equivalent age and four different formula milks. RESULTS: The formula milk for premature infants was the most similar to breast milk of preterm babies. Breast milk showed higher lactose concentrations than formula milk, that conversely presented higher galactose 1-phosphate and maltose concentrations. Mother's milk of very preterm babies (23-25 wks of GA) showed a different metabolic profile from preterm infants ≥29 wks of GA with a subsequent trend to similarity around the 30th week of post-natal age. Breast milk from preterm infants of 29-34 wks, collected up to 40 wks of post-natal age showed a temporal change over the first three weeks of lactation, approaching to zero with the achievement of term age. CONCLUSIONS: Metabolome is a promising tool to study human and artificial milk global metabolic profile.


Subject(s)
Infant Formula/chemistry , Infant, Premature , Metabolomics/methods , Milk, Human/chemistry , Milk/chemistry , Term Birth , Animals , Female , Humans , Infant, Newborn , Principal Component Analysis , Proton Magnetic Resonance Spectroscopy
14.
J Pineal Res ; 57(2): 192-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24980917

ABSTRACT

Conditions that interfere with the endoplasmic reticulum (ER) functions cause accumulation of unfolded proteins in the ER lumen, referred to as ER stress, and activate a homeostatic signaling network known as unfolded protein response (UPR). We have previously shown that in neonatal rats subjected to hypoxia-ischemia (HI), melatonin administration significantly reduces brain damage. This study assessed whether attenuation of ER stress is involved in the neuroprotective effect of melatonin after neonatal HI. We found that the UPR was strongly activated after HI. Melatonin significantly reduced the neuron splicing of XBP-1 mRNA, the increased phosphorylation of eIF2α, and elevated expression of chaperone proteins GRP78 and Hsp70 observed after HI in the brain. CHOP, which plays a convergent role in the UPR, was reduced as well. Melatonin also completely prevented the depletion of SIRT-1 induced by HI, and this effect was observed in the same neurons that over-express CHOP. These results demonstrate that melatonin reduces ER stress induced by neonatal HI and preserves SIRT-1 expression, suggesting that SIRT-1, due to its action in the modulation of a wide variety of signaling pathways involved in neuroprotection, may play a key role in the reduction of ER stress and neuroprotection observed after melatonin.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Hypoxia-Ischemia, Brain/metabolism , Melatonin/pharmacology , Neurons/drug effects , Neurons/metabolism , Sirtuin 1/metabolism , Animals , Animals, Newborn , Rats
15.
Exp Neurol ; 255: 103-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24631374

ABSTRACT

The endoplasmic reticulum (ER) stress can result from several pathological conditions that perturb ER homeostasis and is characterized by accumulation of unfolded proteins in the ER lumen. To cope with ER stress, cells activate the unfolded protein response (UPR), a protein quality control mechanism aimed at restoring homeostasis. The present study was undertaken to characterize the UPR after neonatal hypoxia/ischemia (HI) and its crosstalk with autophagy. After HI, there was a significant increase of GRP78 and Hsp70 expression, phosphorylation of eIF2α, Xbp-1 mRNA splicing and CHOP expression, revealing severe ER stress and UPR. Increasing autophagy with rapamycin (Rap) significantly reduced the UPR. Rap did not further increase the eIF2α phosphorylation and p70S6 kinase (p70S6K) inactivation induced by HI. After autophagy activation, however, there was a clear co-localization between monodansylcadaverine (MDC)-positive autophagosome-like structures and the ribosomal protein S6 (RPS6), indicating the presence of ribosomes in autophagosomes (ribophagy). We found that the autophagy inhibitor 3-methyladenine administered after Rap treatment completely reverted the increased phosphorylation of eIF2α and p70S6K inactivation, and blocked the formation of autophagosome-like structures restoring the UPR. These results demonstrate that the UPR is strongly activated after neonatal HI. Over-activation of autophagy significantly reduces this response, highlighting the relevance of the cross-talk between ER and the autophagy machinery in this important pathological condition. Furthermore, the presence of ribosome subunits in autophagosome-like structures suggests that increased ribosome turnover through autophagy (ribophagy) may represent an additional mechanism involved in the neuroprotective effect observed after autophagy over-activation.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum Stress/physiology , Hypoxia-Ischemia, Brain/metabolism , Protein Biosynthesis/physiology , Unfolded Protein Response/physiology , Animals , Cell Count , Endoplasmic Reticulum/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Rats , Rats, Sprague-Dawley
16.
Tumori ; 100(6): 590-9, 2014.
Article in English | MEDLINE | ID: mdl-25688491

ABSTRACT

Oxidative stress plays a key role in carcinogenesis. Oxidative damage to cell components can lead to the initiation, promotion and progression of cancer. Oxidative stress is also a distinctive sign in several genetic disorders characterized by a cancer predisposition such as ataxia-telangiectasia, Fanconi anemia, Down syndrome, Beckwith-Wiedemann syndrome and Costello syndrome. Taking into account the link between oxidative stress and cancer, the capacity of antioxidant agents to prevent or delay neoplastic development has been tested in various studies, both in vitro and in vivo, with interesting and promising results. In recent years, research has been conducted into the molecular mechanisms linking oxidative stress to the pathogenesis of the genetic syndromes we consider in this review, with the resulting identification of possible new therapeutic targets. The aim of this review is to focus on the oxidative mechanisms intervening in carcinogenesis in cancer-prone genetic disorders and to analyze the current status and future prospects of antioxidants.


Subject(s)
Antioxidants/therapeutic use , Genetic Diseases, Inborn/complications , Genetic Diseases, Inborn/metabolism , Neoplasms/metabolism , Neoplasms/prevention & control , Oxidative Stress , Ataxia Telangiectasia/complications , Ataxia Telangiectasia/metabolism , Beckwith-Wiedemann Syndrome/complications , Beckwith-Wiedemann Syndrome/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Costello Syndrome/complications , Costello Syndrome/metabolism , Down Syndrome/complications , Down Syndrome/metabolism , Fanconi Anemia/complications , Fanconi Anemia/metabolism , Humans , Infant , Neoplasms/genetics , Oxidative Stress/drug effects , Risk
17.
BMJ Case Rep ; 20132013 Mar 07.
Article in English | MEDLINE | ID: mdl-23475990

ABSTRACT

We describe a rare case of cardiac multichamber thrombosis in a young woman admitted with heart failure and atrial fibrillation, who was later found to have reversible postpartum thyrotoxic cardiomyopathy. A transoesophageal echocardiogram demonstrated a patent foramen ovalis with an over-riding thrombus. The clinical scenario was complicated by a cardioembolic stroke upon spontaneous restoration of sinus rhythm.


Subject(s)
Atrial Fibrillation/etiology , Coronary Thrombosis/etiology , Foramen Ovale, Patent/complications , Stroke/etiology , Thyrotoxicosis/complications , Adult , Anticoagulants/therapeutic use , Atrial Fibrillation/diagnosis , Atrial Fibrillation/drug therapy , Coronary Thrombosis/diagnosis , Coronary Thrombosis/drug therapy , Diagnosis, Differential , Echocardiography, Transesophageal , Electrocardiography , Female , Foramen Ovale, Patent/diagnosis , Humans , Postpartum Period , Tomography, X-Ray Computed
18.
Anticancer Res ; 33(2): 691-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23393369

ABSTRACT

BACKGROUND: Costello syndrome is a rare genetic condition characterized by coarse facies, short stature, loose folds of skin especially on hands and feet, severe feeding difficulties and failure to thrive. Other features include cardiac anomalies, developmental disability and increased risk of neoplasms. Given the link between oxidative stress (OS) and carcinogenesis, we tested the hypothesis that OS occurs in this syndrome, supposing its role both in cancer development and in other clinical features. PATIENTS AND METHODS: We describe four cases with Costello syndrome in which we verified the presence of OS by measuring a redox biomarker profile including total hydroperoxides, non-protein-bound iron, advanced oxidation protein products, thyols, carbonyl groups and isoprostanes. Thus, we introduced an antioxidant agent, namely potassium ascorbate with ribose (PAR) into the therapy and monitored the redox profile every three months to verify its efficacy. RESULTS: A progressive decrease in OS biomarkers occurred, together with an improvement in the clinical features of the patients. CONCLUSION: OS was proven in all four cases of Costello syndrome. The antioxidant therapy with PAR demonstrated positive effects. These promising results need further research to confirm the relevance of OS and the efficacy of PAR therapy in Costello syndrome.


Subject(s)
Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Costello Syndrome/drug therapy , Costello Syndrome/physiopathology , Oxidative Stress , Costello Syndrome/metabolism , Female , Humans , Infant, Newborn , Male , Oxidation-Reduction , Ribose/therapeutic use
19.
Free Radic Biol Med ; 53(5): 1061-7, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22842050

ABSTRACT

Isoprostanes, neuroprostanes, isofurans, and neurofurans have all become attractive biomarkers of oxidative damage and lipid peroxidation in brain tissue. Asphyxia and subsequent reoxygenation cause a burst of oxygen free radicals. Isoprostanes and isofurans are generated by free radical attacks of esterified arachidonic acid. Neuroprostanes and neurofurans are derived from the peroxidation of docosahexanoic acid, which is abundant in neurons and could therefore more selectively represent oxidative brain injury. Newborn piglets (age 12-36 h) underwent hypoxia until the base excess reached -20 mmol/L or the mean arterial blood pressure dropped below 15 mm Hg. They were randomly assigned to receive resuscitation with 21, 40, or 100% oxygen for 30 min and then ventilation with air. The levels of isoprostanes, isofurans, neuroprostanes, and neurofurans were determined in brain tissue (ng/g) isolated from the prefrontal cortex using gas chromatography-mass spectrometry (GC/MS) with negative ion chemical ionization (NICI) techniques. A control group underwent the same procedures and observations but was not submitted to hypoxia or hyperoxia. Hypoxia and reoxygenation significantly increased the levels of isoprostanes, isofurans, neuroprostanes, and neurofurans in the cerebral cortex. Nine hours after resuscitation with 100% oxygen for 30 min, there was nearly a 4-fold increase in the levels of isoprostanes and isofurans compared to the control group (P=0.007 and P=0.001) and more than a 2-fold increase in neuroprostane levels (P=0.002). The levels of neuroprostanes and neurofurans were significantly higher in the piglets that were resuscitated with supplementary oxygen (40 and 100%) compared to the group treated with air (21%). The significance levels of the observed differences in neuroprostanes for the 21% vs 40% comparison and the 21% vs 100% comparison were P<0.001 and P=0.001, respectively. For neurofurans, the P values of the 21% vs 40% comparison and the 21% vs 100% comparison were P=0.036 and P=0.025, respectively. Supplementary oxygen used for the resuscitation of newborns increases lipid peroxidation in brain cortical neurons, a result that is indicative of oxidative brain damage. These novel findings provide new knowledge regarding the relationships between oxidative brain injury and resuscitation with oxygen.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/injuries , Oxidative Stress/drug effects , Oxygen/pharmacology , Animals , Cerebral Cortex/metabolism , Female , Furans/analysis , Hyperoxia/metabolism , Hypoxia/metabolism , Isoprostanes/analysis , Male , Neuroprostanes/analysis , Oxygen/administration & dosage , Swine
20.
Early Hum Dev ; 88(8): 707-10, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22421197

ABSTRACT

BACKGROUND: During permanence in most incubators, newborns are very close to the electric engine, which represents a source of electromagnetic fields (EMF). Previous studies demonstrated a decrease in melatonin production in adults and animals exposed to EMF. AIMS: To assess melatonin production in a group of newborns exposed to EMF, and to evaluate whether removing the babies from the source of MF can affect melatonin production. STUDY DESIGN AND SUBJECTS: We have recruited 28 babies (study group), who had spent at least 48 h in incubator where we had previously assessed the presence of significant EMF. We have measured their mean 6-hydroxy-melatonin-sulfate (6OHMS) urine excretion at the end of their permanence in the incubators, and compared it with their mean 6OHMS excretion after having been put in cribs, where EMF are below the detectable limit (<0.1mG). We have also measured urine 6OHMS twice, with an interval of 48h, in a control group of 27 babies who were not exposed to EMF during both samples. RESULTS: Mean 6OHMS/cr values were respectively 5.34±4.6 and 7.68±5.1ng/mg (p=0.026) when babies were exposed to EMF in incubators, and after having been put in the crib. In the control group, mean 6OHMS/cr values in the first and in the second sample were respectively 5.91±5.41 vs 6.17±3.94ng/mg (p=0.679). CONCLUSIONS: The transitory increase in melatonin production soon after removing newborns from incubators demonstrates a possible influence of EMF on melatonin production in newborns. Further studies are needed to confirm these data.


Subject(s)
Electromagnetic Fields/adverse effects , Incubators, Infant/adverse effects , Infant, Newborn/urine , Melatonin/analogs & derivatives , Female , Humans , Male , Melatonin/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...