Skip to content
Publicly Available Published by De Gruyter December 19, 2019

Early predictors of perinatal brain damage: the role of neurobiomarkers

  • Iliana Bersani , Francesca Pluchinotta , Andrea Dotta , Immacolata Savarese , Francesca Campi , Cinzia Auriti , Natalia Chuklantseva , Fiammetta Piersigilli , Francesca Gazzolo , Alessandro Varrica , Angela Satriano and Diego Gazzolo EMAIL logo

Abstract

The early detection of perinatal brain damage in preterm and term newborns (i.e. intraventricular hemorrhage, periventricular leukomalacia and perinatal asphyxia) still constitute an unsolved issue. To date, despite technological improvement in standard perinatal monitoring procedures, decreasing the incidence of perinatal mortality, the perinatal morbidity pattern has a flat trend. Against this background, the measurement of brain constituents could be particularly useful in the early detection of cases at risk for short-/long-term brain injury. On this scenario, the main European and US international health-care institutions promoted perinatal clinical and experimental neuroprotection research projects aimed at validating and including a panel of biomarkers in the clinical guidelines. Although this is a promising attempt, there are several limitations that do not allow biomarkers to be included in standard monitoring procedures. The main limitations are: (i) the heterogeneity of neurological complications in the perinatal period, (ii) the small cohort sizes, (iii) the lack of multicenter investigations, (iv) the different techniques for neurobiomarkers assessment, (iv) the lack of consensus for the validation of assays in biological fluids such as urine and saliva, and (v), the lack of reference curves according to measurement technique and biological fluid. In the present review we offer an up-to-date overview of the most promising developments in the use of biomarkers in the perinatal period such as calcium binding proteins (S100B protein), vasoactive agents (adrenomedullin), brain biomarkers (activin A, neuron specific enolase, glial fibrillary acidic protein, ubiquitin carboxyl-terminal hydrolase-L1) and oxidative stress markers.

Introduction

There has been a dramatic increase in recent years in the number of studies focusing on neurobiomarkers (NBs) of central nervous system (CNS) injury [1], [2], [3], [4], [5], [6], [7], [8], [9], [10], [11]. The optimal NB should be reliable, easily and harmless to collect, reproducible and able to guide caregivers in daily practice. The Food and Drug Administration (FDA), the European Medicines Agency (EMA) and the National Institutes of Health (NIH) are supportive of research into biomarkers in order to: (i) improve the early identification of cases at risk, (ii) promote specific preventive or therapeutic treatments [2], and (iii) provide health care professionals with a new tool, as well as to set the stage for a more modernized standard of care for the testing of suspect cases (Table 1).

Table 1:

Criteria for an NB inclusion in clinical guidelines according to the FDA and EMA statements.

Optimality items for an NB according to the FDA and EMA
1. Alternative and direct indicator of CNS damage when clinical and radiologic assessments are still silent
2. Early predictor of degree and location of injury
3. Indicator of the extent of brain lesion
4. Monitor the progression of disease
5. Well studied in the pediatric population
6. Measurable by available commercially kits worldwide with good reproducibility
7. Presence of reference range for the pediatric population
8. Assessment in different biological fluids (urine, blood, CSF, amniotic fluid, saliva, milk)
  1. CSF, cerebrospinal fluid; CNS, central nervous system; EMA, European Medicines Agency; FDA, Food and Drug Administration; NB, neurobiomarker.

The optimal NB should monitor disease progression by longitudinal assessments and possibly correlate with standard procedures such as cerebral ultrasound (CUS) and magnetic resonance imaging (MRI) to assess the entity of brain injury. The ranges of normality of the ideal NB should be available for both healthy term and preterm neonates, and possibly identifiable in different biologic fluids (amniotic; cerebrospinal fluid [CSF]; blood; urine; saliva; milk). However, to date only a few NBs meet these criteria for routine use [2].

The present review offers an up-to-date overview of the most promising developments regarding the use of NBs closer to FDA and EMA criteria, such as S100B protein, adrenomedullin (AM), activin A (AcA), neuron specific enolase (NSE), oxidative stress markers (OSM), glial fibrillary acidic protein (GFAP) and ubiquitin carboxyl-terminal hydrolase-L1 (UCH-L1), in the perinatal period. This field is particularly challenging due to the heterogeneity of neurological complications which may complicate the early life periods.

S100B

S100B protein and its heterodimeric form ββ is highly specific for the CNS being expressed mostly in glial cells but also detectable in neuronal subpopulations [12], [13], [14]. S100B has a half-life of 1-h and is eliminated mostly by the kidneys (98%) [15]. S100B has been detected in different biological fluids such as amniotic, CSF, blood, urine, saliva and milk [16], [17], [18], [19], [20], [21], [22], [23]. It has neurotrophic effects at physiologic levels (nanomolar), but becomes neurotoxic at high concentrations (micromolar) [24], [25], [26], [27].

CSF

The first studies investigating S100B as a perinatal NB were performed using CSF. Whitelaw et al. showed that S100B was higher than controls in preterm newborns with post-hemorrhagic ventricular dilatation [28], [29].

In term neonates, Blennow et al. found that S100B in the CSF of term newborns, complicated by perinatal asphyxia (PA), correlated significantly with neurological impairment at 1-year of age, or death before that time [30]. These results were therefore comparable to those recorded in preterm newborns [28], [29]. However, because of ethical and medical issues and the impossibility of longitudinal monitoring, the assessment of S100B in CSF was progressively abandoned.

Blood

Following previous observations in CSF, S100B was measured in blood. Among a panel of biomarkers, S100B had the highest sensitivity in cord blood for the prediction of brain injury in preterm infants. S100B at a cut-off of 1.07 μg/L achieved a specificity of 53% and a sensitivity of 93% as a predictor of brain damage in preterm infants [23] (Table 2).

Table 2:

Currently available studies providing data about NBs availability as predictors of brain damage and/or ominous outcome.

Biomarker Fluid Disease Cut-off PPV, % NPV, % Specificity, % Sensitivity, % Reference
S100B CB PA-HIE 8.5 μg/L 71 90 90 71 [31]
CB PN, BD 1.07 μg/L NA NA 52.9 95.3 [23]
PB PA-HIE 1.6 ng/mL NA NA 91 40 [32]
U PN-IVH 0.70 μg/L NA NA 100 100 [33]
U IUGR-BD 7.37 MoM 100.7a 0.05a 99.1 95 [34]
U PN-D 12.93 MoM 78.6 100 97.8 100 [35]
U PA-BD 0.28 μg/L 46.2 100 87.3 100 [36]
U PA-HIE 0.41 μg/L 80.8 97.8 94.6 91.3 [37]
U TN-D 1.0 μg/L 100 100 100 100 [38]
U TN-D 1.11 μg/L NA NA 60 100 [39]
U TN-BD 0.66 μg/L NA NA 70 83 [39]
S CHD-BD 3.25 MoM 100 100 100 100 [40]
AM PB CHD-BD 17.4 ng/L 3.7a 0.0a 73 100 [41]
AB CHD-LCOS 27.0 pg/L 39.1 100 64.1 100 [42]
AcA CSF PA-HIE 1.3 ng/L 100 0 100 100 [43]
CB PN-IVH 0.8 μg/L 79 100 93 100 [44]
CB PN-BD 321 ng/L NA NA 92 86 [23]
PB PA-HIE 0.66 ng/L 27.69a 0.069a 96 93 [45]
PB CHD-BD 0.94 ng/L 100 0 100 100 [46]
U PN-IVH 0.08 ng/L 45.8 93.4 84.5 68.7 [47]
U PA-HIE 0.08 ng/L NA NA 100 83 [48]
NSE CB PA-HIE 44.0 μg/L 46 93 83 68 [31]
PB PA-HIE 40.0 μg/L 51 89 70 79 [49]
PB PA-BD 45.4 μg/L 39 95 70 84 [49]
PB PA-HIE 81.0 ng/mL NA NA 83 71 [32]
OS CB TN-PN-BD 15.2 μmol/L NA NA 100 100 [50]
G-FAP CB PN-PVL-WM >0.04 ng/mL NA NA 91.2 52.4 [51]
PB PA-HIE >0.08 ng/mL NA NA 100 100 [52]
PB PA, BD, D 0.2 ng/mL 78 90 82 87.5 [53]
PB PA, HIE 0.07 ng/mL NA NA 78 77 [54]
UCH-L1 PB PA, BD, D 13.8 ng/mL 78 90 100 75 [53]
PB PA, HIE 18.0 ng/mL NA NA NA NA [55]
PB PA, HIE, BD 28.0 ng/mL NA NA 95 NA [56]
  1. AcA, activin A; AM, adrenomedullin; BD, brain damage; CSF, cerebrospinal fluid; BF, biologial fluid; CB, cord blood; D, early neonatal death; G-FAP, glial fibrillary acid protein; HIE, hypoxic ischemic encephalopathy; IVH, intraventricular haemorrhage; MB, maternal blood; NPV, negative predictive value; NSE, neuron specific enolase; OSM, oxidative stress markers; PB, peripheral blood; PN, preterm neonate; PPV, positive predictive value; PVL, periventricular malacia; S, saliva; TN, term neonate; UCH-L1, ubiquitin carboxyl-terminal hydrolase L1; U, urine; WM, white matter injury. aLikelihood ratio (positive/negative).

Conversely, Costantine et al. showed that the predictive value of S100B for the development of cerebral palsy was weaker after adjusting for gestational age (GA) and perinatal treatment such as magnesium sulfate [57].

Increased S100B peripheral blood levels were also observed in: (i) 24 preterm newborns complicated by intraventricular hemorrhage (IVH) 48 h before the development of any clinical, laboratory or ultrasonographic sign of hemorrhage. A correlation between S100B and the extent of IVH was also reported [58]; (ii) in preterm newborns complicated by intrauterine growth restriction (IUGR), presumably as a consequence of the hemodynamic re-arrangement [59], [60], [61].

In term newborns, increased concentrations of serum S100B have been observed in PA infants complicated by hypoxic-ischemic encephalopathy (HIE). Nagdyman et al. measured cord blood concentrations of a panel of NBs including S100B, in a cohort of 49 term newborns. They found higher S100B in PA infants with moderate-severe HIE at 2 h after birth. Moreover, a combination of S100B (cut-off value: 8.5 μg/L) and CK-BB (cut-off value: 18.8 UI/L) as early as 2 h after birth had the highest sensitivity (83%) and specificity (95%) in predicting moderate and severe HIE [31].

S100B cord blood levels were also assessed in 13 term infants with stage II–III HIE and compared to 21 healthy controls. Higher S100B concentrations were associated with cord blood acidosis, aEEG pattern severity, HIE severity, and correlated with neurodevelopmental sequelae at 6-year follow-up and death) [62]. In contrast, Summanen et al. found cord blood S100B to be a poor NB of PA in term newborns [63] (Table 2).

Lastly, in a cohort of 100 term newborns of whom 20 complicated by PA-HIE and intracranial hemorrhage (ICH), S100B was significantly higher in the PA-ICH group than in those who did not develop ICH and in controls [64].

Urine

In preterm infants, urine S100B levels were longitudinally measured in the first 72 h from birth in a cohort of 36 cases, of whom 18 developed IVH. Increased S100B at birth was detected 72 h before the development of any clinical, laboratory or ultrasonographic sign of IVH. At a cut-off of 0.70 μg/L, the sensitivity and specificity of urinary S100B as a diagnostic test were 100%. S100B also correlated with the extent of IVH by means of longitudinal CUS recordings [33] (Table 2).

In IUGR infants, S100B was measured longitudinally in the first 7 days from birth in a cohort of 84 infants, of whom 42 were growth restricted. S100B was higher at all monitoring time-points in the IUGR newborns showing an abnormal neurological outcome than in those who did not and in controls. At a cut-off of 7.37 multiples of median (MoM) at first urination, S100B achieved a sensitivity of 95% and a specificity of 99.1% as a single marker for predicting an adverse short-term neurological outcome [34] (Table 2).

S100B was also longitudinally measured (in the first 96 h from birth) in 165 preterm newborns of whom 11 suffered early neonatal death, 121 displayed no overt neurologic syndrome, and 33 suffered neonatal hypoxia and IVH but no ominous outcome. S100B concentration was higher at all monitoring time-points in preterm newborns who later died than in the other studied groups. At a cut-off of 12.93 MoM at first void, sensitivity was 100%, specificity 97.8%, and positive (PPV) and negative predictive values (NPV) 78.6% and 100%, respectively, for predicting early post-natal death [35] (Table 2).

In a cohort of 277 late preterm infants, S100B was found to be gender- and GA-dependent [65].

In PA infants, urine S100B was longitudinally measured in the first 72 h from birth in a cohort of 134 term newborns, of whom 38 were complicated by PA-HIE with normal (n=20) or abnormal (n=18) 1 year neurological follow-up and 96 controls. S100B was higher in PA infants developing an abnormal neurological outcome than in normal PA infants and controls. At a cut-off value of 0.28 μg/L at first urination, S100B achieved a sensitivity of 100% and a specificity of 87.3% for predicting an adverse neurological follow-up [36]. The same authors also found higher S100B in severe PA-HIE infants than in mild PA-HIE infants and controls. An S100B concentration cut-off of 0.41 μg/L at first urination had a sensitivity of 91.3%, a specificity of 94.6%, a PPV of 80.8% and an NPV of 97.8% as a predictor of HIE [37] (Table 2).

As for preterm newborns, the diagnostic accuracy of S100B in predicting early postnatal death was investigated in a cohort of 132 term newborns, of whom 48 had PA-HIE and 12 died within 7 days from birth. Higher S100B was observed in the PA-HIE newborns who died than in the other groups studied. At a cut-off of 1.0 μg/L S100B had a sensitivity/specificity, PPV and NPV of 100% for predicting neonatal death [38] (Table 2).

Finally, Alshweki et al. measured S100B in the urine of 31 PA-HIE newborns with normal (n=13) and abnormal/ominous outcome (n=18) detected by MRI. Higher S100B was observed in infants with unfavorable outcome. An S100B cut-off of 1.11 μg/L had a sensitivity of 100% and a specificity of 60% for the prediction of neonatal death, whilst at a cut-off of 0.66 μg/L S100B had a sensitivity of 83% and a specificity of 70% for the prediction of abnormal neurological outcome [39] (Table 2).

Saliva

Data in the literature show that S100B: (i) is essentially absent from fetal salivary glands [66], (ii) is not produced by salivary glands, and that (iii) saliva concentrations are derived from systemic circulation [67]. A reference curve of S100B in saliva was therefore provided in a cohort of 216 preterm and term newborns. The results showed, as for other biological fluids (cord blood, urine), that S100B was higher in preterm than in term newborns and was GA-dependent [21]. The findings allowed the identification of saliva as offering the least stressful fluid for CNS clinical monitoring in a neonatal intensive care unit (NICU).

In an international multicenter study, recruiting a cohort of 292 term newborns, of whom 48 suffered PA and 244 healthy controls longitudinal S100B saliva concentrations correlated with the presence of neurological abnormalities at 1 year after birth. S100B was higher in PA newborns with poor prognosis than in PA with good prognosis or controls. At a cut-off >3.25 MoM, S100B achieved a sensitivity/specificity of 100% for predicting the occurrence of abnormal MRI patterns and neurological outcome [40] (Table 2).

S100B and perinatal therapeutic strategies

S100B has also been investigated to evaluate the pros and cons of in utero and post-natal therapeutic strategies such as antenatal maternal glucocorticoid (GC) treatment, nitric oxide donor (NO) and selective serotonin re-uptake inhibitors (SSRI) [68], [69], [70].

GC supplementation is widely used for the prevention of lung immaturity, but no conclusive data on its possible side-effects on other organs, including the CNS, are available [71]. In this regard, preterm newborns, antenatally GC-treated showed lower longitudinal urinary S100B levels (in the first 120 h from birth) than in controls [68]. The findings are supportive of an inhibited release of S100B as a neurotrophic factor, offering additional evidence in the debate on the side-effects on CNS development of GC. Notably, Sannia et al. found that S100B levels, measured in the first 72 h changed in a GC dose-dependent manner in preterm newborns whose mothers received a complete course of GC than in infants whose mothers received half a course or no GC [72]. In recent decades, the effects of NO in IUGR fetuses with placental insufficiency have been investigated in a randomized controlled multicenter trial admitting to the study 48 IUGR pregnancies treated with either placebo (n=25) or a transdermal glyceryl-trinitrate patch (5 mg every 16 h daily) until delivery (n=23). S100B cord blood levels were lower in NO-treated pregnancies than in the placebo group and an improved neurological and respiratory outcome was also observed [69].

Finally, the use of SSRI during pregnancy is increasing both in Europe (2%–3%) and in the USA (8%–10%) [73]. This trend has evolved without any solid evidence on the safety or efficacy of this approach, and treatment is recommended for pregnant women with depression despite the potential side-effects on the fetus and newborn [73], [74], [75], [76]. In light of this, S100B was assessed in 306 pregnant women, of whom 75 were SSRI-treated, and 231 healthy controls, and their newborns. The results showed higher S100B in maternal-fetal-neonatal fluids in the SSRI-treated group, particularly in infants with 7-day adverse neurological outcome [70].

S100B and hypothermia

Today, hypothermia (HT) is an accepted standard of care therapeutic strategy for term newborns complicated by PA-HIE [77], [78]. Longitudinal blood levels of a panel of NBs, including S100B, were measured during HT in a cohort of 83 PA-HIE infants. S100B increased during HT and was associated with adverse neurodevelopmental outcome at 15 months of age [79]. Similarly, Massaro et al. correlated S100B with cerebral MRI patterns at 14 day neurological follow-up in 75 HT-treated PA-HIE infants. They found higher S100B in term newborns with unfavorable outcome and pathological MRI patterns. S100B at a cut-off of 1.6 ng/mL reached a specificity and a sensitivity of 91% and 40%, respectively, as a diagnostic test of brain injury [32] (Table 2). These data partly agreed with those of Roka et al. [80], who found, in a cohort of 24 HT (n=13) or non-HT-treated (n=11) PA-HIE infants, elevated S100B in both HT and no-HT groups.

Recently, the potential neuroprotective effects of erythropoietin were evaluated in 50 HT-treated PA-HIE infants, using a panel of NBs including S100B. The protein increased in infants showing pathological cerebral MRI patterns. No significant effects of erythropoietin treatment on NB levels were detectable [81].

HT can be performed as selective head cooling or whole-body cooling. Celik et al. compared the efficacy of the two HT procedures using a panel of NBs including S100B in 21 PA-HIE infants. Neurological follow-up was set at 1 year after birth. The results showed no differences in NBs between the two HT methods [82].

AM

AM, first isolated from human pheochromocytome, is a vasodilator peptide detectable in multiple tissues [83], [84], [85]. AM’s half-life is 2 h and it is eliminated mostly by the kidneys [86]. Both in vitro and in vivo, AM: (i) influences local and systemic blood pressure [87], (ii) plays a special role in the regulation of blood flow [88], (iii) has a role in cardiovascular adaptation after birth [89], [90], [91], (iv) is released after stressful events such as hypoxia and PA [91], [92], and (v) as a neuropeptide, regulates cerebral blood flow [93], [94].

CSF

To the best of our knowledge, no studies have measured AM in the CSF of preterm newborns. In term newborns and children, higher levels of AM were found in the CSF after traumatic brain injury as an endogenous response to cerebral hypoperfusion [93].

Blood

In cord blood, AM was measured in IUGR (n=16) and non-IUGR (n=16) infants. Higher AM was found in IUGR infants than in controls, suggesting that AM acts as a compensatory mechanism in response to chronic hypoxia and its hemodynamic re-arrangement characterized by the brain-sparing effect [95].

In venous blood of preterm newborns developing IVH (n=24) and controls (n=48), higher levels of AM were found within 6 h from birth, suggesting the involvement of AM in the loss of cerebral vascular regulation secondary to a hypoxic insult [96].

In term newborns, Kamata et al. found that cord AM levels in the umbilical artery and vein were higher in infants with persistent pulmonary hypertension (n=15) than in controls (n=8) [97]. In PA newborns developing IVH (n=20) and in controls (n=20), higher AM was observed in PA-IVH infants, suggesting its involvement in the loss of cerebral vascular regulation due to HI [98].

AM has also been measured in a cohort of 80 infants complicated by significant hyperbilirubinemia (n=40) or non-significant hyperbilirubinemia (n=40). Higher AM was observed in infants with significant jaundice, suggesting its involvement in adverse effects and neuronal injury steps of hyperbilirubinemia [99].

In congenital heart disease (CHD) term newborns complicated (n=40) or not (n=10) by adverse 1-year neurological outcome, decreased AM levels were observed in neurologically abnormal infants. At a cut-off of 17.4 ng/L, AM as a predictor of neurological abnormalities achieved a sensitivity of 100%, a specificity of 73.0%, and positive (PLHr) and negative (NLHr) likelihood ratios of 3.7 and 0.0, respectively [41]. Furthermore, in CHD infants complicated (n=9) or not (n=48) by intra-operative low cardiac output syndrome (LCOS), Abella et al. found lower AM in LCOS infants. At a 27 pg/L cut-off, AM achieved a sensitivity of 100%, a specificity of 64.1%, a PPV of 39.1% and a NPV of 100.0% for the prediction of LCOS [42] (Table 2).

AcA

AcA is a member of the TGF-beta superfamily involved in the regulation of a variety of functions, including cell proliferation, differentiation, bone remodeling, hematopoiesis, wound healing and apoptosis [100], [101]. AcA promotes neuronal differentiation, and increased AcA levels in biological fluids have been found under HI conditions, as a response to brain injury, and as a local mediator of angiogenesis during the repair process [102], [103].

AcA also has a beneficial role in neuronal recovery, supporting the survival of neurogenic cell lines and retinal neurons [104], and offering protection against neurotoxins of different origins [105]. In animal models, AcA enhanced the survival of embryonic hippocampal neurons [106], to decrease ischemic brain injury and rescue striatal neurons against neurotoxic damage [107].

CSF

To the best of our knowledge, no studies have measured AcA in the CSF of preterm newborns.

AcA was assessed within 24 h in the CSF of 74 term newborns, of whom 30 were PA newborns and 44 controls. Higher AcA was found in infants affected by severe HIE than in moderate HIE and controls. At the cut-off of 1.3 ng/L, AcA reached a sensitivity and specificity of 100%, a PPV of 100% and an NPV of 0% as a predictor of HIE [43] (Table 2).

Blood

Florio et al. measured AcA in cord blood of hypoxic (n=26) and non-hypoxic (n=24) preterm newborns. Higher AcA was found in hypoxic preterm newborns than in controls. The main explanations is that AcA can act as a biomarker of hypoxia and/or be an expression of neuroprotective process activation [108]. The same authors measured cord blood AcA levels in 50 high-risk pregnancies and 40 controls. Higher AcA was observed in high-risk newborns and correlated with the occurrence of the brain-sparing effect and the length of hospital stays [109].

In preterm newborns developing (n=11) or not (n=42) IVH, AcA was measured in arterial cord blood soon after birth (2-h). Higher AcA was observed in IVH preterm newborns than in controls. At a cut-off of 0.8 μg/L, AcA has a sensitivity of 100%, a specificity of 93%, a PPV of 79% and an NPV of 100% as a predictor of IVH [44] (Table 2).

In a cohort of 130 preterm infants, Lu et al. measured amniotic and cord blood levels of a panel of biomarkers (AcA, interleukin-1β, interleukin-6, interleukin-8, tumor necrosis factor-α, granulocyte colony-stimulating factor, monocyte chemotactic protein-1, soluble intercellular adhesion molecule-1, S100B) among which AcA was the best predictor of long-term brain injury. AcA at a cut-off of 321 ng/L achieved a specificity of 92% and a sensitivity of 86% as a predictor of brain damage in preterm infants [23] (Table 2).

AcA concentrations in cord blood were assessed in 26 hypoxic infants and 60 controls. AcA was higher in hypoxic preterm newborns in a gender-dependent manner [110].

In blood, AcA was longitudinally assessed in 35 PA-HIE newborns and 70 controls. AcA was higher in infants affected by severe HIE than in moderate-HIE infants and controls. At the cut-off of 0.66 ng/L, AcA achieved, as a predictor of HIE, a sensitivity of 93%, a specificity of 96%, a PLHr of 27.69 and NLHr of 0.069 [45] (Table 2).

AcA was measured in the perioperative period in 45 CHD infants, of whom 36 were without overt neurologic injury and nine had a neurologic injury at 7 days after surgery. Higher AcA in the perioperative period was found in neurologically abnormal than in neurologically normal infants. At a cut-off of 0.94 ng/L, AcA had a sensitivity and specificity of 100% for predicting perioperative neurological abnormalities [46] (Table 2).

Urine

In preterm newborns developing (n=20) or not (n=80) IVH, AcA was longitudinally measured in urine soon after birth (2-h). Higher AcA was observed in IVH preterm newborns than in controls. At a cut-off of 0.08 ng/L, at the first void, AcA had a sensitivity of 68.7%, a specificity of 84.5%, PPV and NPV 46%–93%, respectively, as a predictor of IVH [47] (Table 2).

In urine, AcA was longitudinally assessed in 30 PA-HIE infants and 30 controls. Higher AcA was found in infants affected by severe-HIE than in moderate-HIE and controls. At a cut-off of 0.08 μg/L, AcA achieved, as a predictor of HIE, a sensitivity of 83% and a specificity of 100% [48] (Table 2).

AcA and perinatal therapeutic strategies

AcA concentrations in maternal and fetal biological fluids were assessed in 24 mothers antenatally treated with SSRI and in 24 controls. Higher AcA was observed in maternal and fetal biological fluids in SSRI-treated subjects than in controls. The authors suggested that AcA may play a key role as a new therapeutic option and/or marker of maternal/fetal CNS stress in pregnant women with depression [111].

NSE

NSE, a glycolytic enzyme detected at high concentrations in neuronal cytoplasm [112], represents a late marker of neural differentiation and maturation. NSE is released into the extracellular space in cases of cell death [113].

CSF

In children, NSE increases in both CSF and blood after impairment of the blood-brain barrier and brain injury [114], [115], [116]. Few studies have investigated the role of NSE in neonates.

Increased NSE in CSF was observed in PA-HIE term newborns compared with controls [117], [118], [119].

Blood

Costantine et al. performed a multicenter randomized trial of magnesium sulfate administration vs. placebo administration to prevent cerebral palsy development or death in preterm infants. The results showed that NSE cord blood levels were unable to differentiate preterm infants who developed cerebral palsy (n=16) or died within 1 year of age (n=25) from those who did not [57].

In the blood of 18 CHD infants undergoing open-heart surgery and cardiopulmonary bypass (CPB), higher NSE was observed in the perioperative period in infants with adverse neurological outcome [120].

In 30 PA-HIE term newborns, cord blood NSE levels were higher than in the 30 controls [121]. In this regard, Nagdyman et al. found higher NSE in PA infants with moderate-severe HIE (n=7) at 2 h after birth than mild HIE (n=22) and controls (n=20). An NSE cut-off at 2 h of 44.0 μg/L achieved a sensitivity of 68%, a specificity of 83%, a PPV and NPV of 46%–93% in predicting moderate-severe HIE [31] (Table 2).

Higher blood NSE concentrations were observed in 43 PA-HIE infants than in controls and correlated with the severity of HIE. At a cut-off of 40.0 μg/L, NSE had a sensitivity of 79% and a specificity of 70% as a predictor of HIE. Indeed, at a cut-off of 45.4 μg/L, NSE has a sensitivity of 84% and a specificity of 70% as a predictor of poor outcome [49] (Table 2).

Finally, HT-treated PA-HIE infants showed higher levels of NSE in term newborns who developed MRI and clinical patterns suggesting brain injury [32], [80]. In particular, Massaro et al. found higher NSE concentrations in term newborns with unfavorable outcome and pathological MRI patterns. At a cut-off of 81 ng/mL, NSE reached a specificity and a sensitivity of 83% and 61%, respectively, as biomarker of brain injury [32] (Table 2). Similarly, Roka et al. found, in a cohort of 24 HT (n=13) or non-HT-treated (n=11) PA-HIE infants, higher NSE values in infants who died or developed severe neurological impairment [80].

OSM

It is known that the antioxidant systems of fetuses and newborns are immature, and therefore exposed to the damaging effects of oxidative stress [122]. Free radicals are highly reactive substances involved in self-amplified chain reactions leading to cell death or apoptosis. Once the balance between the production of antioxidant enzymes and of free radicals changes in favor of the latter, oxidative stress damage may occur [123]. Oxidative stress is thus involved in the pathogenesis of many fetal and neonatal diseases, mainly triggered by hypoxia.

CSF

OSM (8-isoprostane; malondialdehyde [MDA]; protein carbonyl [PC]; chlorotyrosine) were measured in 22 preterm newborns developing white matter injury, 30 term newborns and 17 adults. Higher OSM were observed in preterm newborns than in the other groups studied [124]. The same authors also showed, in a case report, increased levels of the mentioned OSM in a preterm newborn complicated by periventricular leukomalacia (PVL) [125].

No differences were found in CSF OSM (nitric oxide, nitrotyrosine) levels between mild PA-HIE term newborns (n=11) and controls (n=9) [126]. Lower levels of CSF lipid peroxides and antioxidant enzymes (superoxide dismutase [SOD]; glutathione peroxidase, GSHP, MDA) were found in 72 PA-HIE term newborns after high-dose administration of phenobarbital [127]. In animal models, lower MDA levels have been reported after the administration of melatonin [128].

Blood

Buonocore et al. measured plasma non-protein-bound iron (NPBI) levels in the cord blood of a cohort of 384 newborn infants of whom 51 showed an abnormal neurodevelopmental outcome. At a cut-off of 15.2 mmol/L, NPBI reached a sensitivity and specificity of 100% as a diagnostic test of adverse neurodevelopmental outcome (Table 2) [50].

Comporti et al. found, in a cohort of 24 preterm newborns and 27 term newborns, higher F2-isoprostanes in the blood of preterm than term infants in correlation with GA. They suggested the involvement of OSM in the physiopathological changes related to perinatal growth [129]. An identical pattern was observed when NPBI was measured in a cohort of 30 preterm and 29 term newborns [130].

Cord blood OSM were assessed in term newborn (n=28), preterm newborn (n=28) IUGR infants and controls (n=24). The results showed that increased levels of peroxynitrite anion and thiobarbituric acid-reactive substances correlated with the CNS maturity of IUGR infants [131].

OSM (MDA, SOD, catalase, CAT; GSH) were assessed in the cord blood of 20 SGA term newborns and 20 controls. Significant differences were observed between the groups, suggesting that intrauterine malnutrition is associated with increased OSM in SGA term newborns [132]. Furthermore, OSM (MDA, SOD) were measured in SGA infants delivered by cesarean section (n=21) or vaginally (n=21). Results showed that SGA term newborns delivered by caesarean section had insufficient protective mechanisms against increased OSM at birth [133].

Higher longitudinal blood OSM (MDA, PC) were detected in PA-HIE term newborns (n=40) than in controls (n=40). The data correlated with the occurrence of 8 m adverse neurological outcome and early neonatal death [134].

In HT-treated PA-HIE term newborns (n=10) and controls (n=11), blood OSM (total hydroperoxides) were longitudinally measured up to 72 h from birth. Higher OSM were found in HT-treated term newborns, suggesting a partial protective action by HT [135].

Blood OSM (CAT, GSH, nicotinamide-adenine dinucleotide phosphate ratio, MDA) concentrations were also compared in term (n=100) and preterm newborns (n=100). Lower OSM were observed in preterm newborns, suggesting that they are much more exposed to OSM at birth and are susceptible to antioxidant deficiencies [136].

Finally, levels of OSM (xanthine, hydroperoxides, advanced oxidative protein products, glutathione s-transferases) measured in urine have been found to correlate with non-CNS diseases [137], [138].

GFAP

GFAP is a cytoskeletal monomeric filament protein, detectable in the astroglia of the CNS, representing a specific marker of differentiated astrocytes [139], [140]. As GFAP is not routinely secreted in blood but only as a consequence of astrocyte death, it has been investigated as a biomarker for brain injury.

CSF

In the CSF of a cohort of preterm neurologically abnormal (n=10) or normal newborns (n=7) and of term newborns (n=9), increased levels of GFAP were found to correlate with abnormal neurological outcome [141].

Blood

Longitudinal GFAP cord blood levels (0–96 h) were assessed in 21 very low birthweight (VLBW) newborns, affected by PVL and white matter injury (WMI) on CUS at 6 weeks of life, matched for GA with 42 healthy controls. GFAP was higher in damaged infants from 24 to –96 h time-point. A GFAP cut-off >0.04 ng/mL at 24 h time-point achieved a specificity and a sensitivity of 91.2% and 52.4% as predictor of PVL and WMI [51] (Table 2).

In 56 CHD term newborns, blood GFAP levels at birth were superimposable on those measured in 23 controls, and started to change after the transitional phase (>72 h) in parallel with closure of the patent ductus arteriosus [142].

GFAP cord blood levels were investigated in PA-HIE and controversial results have been observed. In particular: (i) no differences between PA-HIE newborns (n=15) and controls (n=31) [143]; (ii) no differences and no correlation with the outcome at 36 months follow-up [144], (iii) higher GFAP from birth to 96-h of life in PA-HIE newborns (n=15) than in controls (n=11) [55], and (iv) increased GFAP at birth in 20 PA-HIE term newborns developing poor neurological outcome at 18 months follow-up [52]. GFAP at a cut-off of >0.08 ng/mL at 12 h achieved a PPV and NPV of 100% and 0%, respectively [52] (Table 2).

In 23 HT-treated PA-HIE infants, GFAP was predictive of brain injury as shown by MRI patterns [145]. In 64 HT-treated PA-HIE infants, Jiang et al. found higher longitudinal GFAP in moderate/severe than in mild PA-HIE cases [54]. Finally, in 20 PA-HIE infants Massaro et al. found increased GFAP at 24–72 h in cases with poor/ominuous outcome. In light of this, GFAP at a off of 0.2 ng/mL achieved a specificity of 90%, a sensitivity of 78% and PPV-NPV of 82%–87%, respectively, as predictor of poor/ominous outcome [53] (Table 2).

More recently, Patil et al. measured GFAP in a cohort of PA infants potentially suitable for HT. They found a significant correlation between increased GFAP levels and the occurrence of abnormal aEEG [146].

UCH-L1

UCH-L1 is a neuron-specific cytoplasmatic enzyme concentrated in the perikarya and dendrites of neurons. Some authors have proposed UCH-L1 as a desirable NB as it is an abundant protein specific to the CNS and resistant to endogenous proteases [147].

CSF

To the best of our knowledge, no studies have measured UCH-L1 in the CSF of preterm and term newborns.

Blood

In blood, increased UCH-L1 levels have been reported as an expression of altered blood-brain barrier permeability [148]. Blood UCH-L1 has also been investigated as a marker of traumatic injury in pediatric patients, with promising results [148], [149].

In 15 PA infants with moderate/severe-HIE and 31 controls, cord blood UCH-L1 has been reported not to differ among studied groups [143]. Furthermore, Jiang et al. found higher UCH-L1 in 31 HT-treated PA-HIE infants than in 34 controls: UCH-L1 also correlated with the degree of HIE [54].

In 20 HT-treated PA-HIE infants, Chalak et al. found no correlation between UCH-L1 and adverse neurological outcome [52]. Conversely, Douglas-Escobar found that UCH-L1 in the cord blood of 16 PA infants was associated with cortical injury and later motor and cognitive development [55]. The same Authors, in a cohort of 14 PA-HIE infants, found a correlation between UCH-L1 and the occurrence of ominous outcome, with a specifity of 95% for a cut-off value of 28.0 ng/mL [56]. Increased UCH-L1 has also been reported by Massaro et al. at different HT monitoring time-points in 20 PA-HIE infants with poor outcome. At a cut-off of 13.8 ng/mL, UCH-L1 reached a specificity of 100% and a sensitivity of 75% as a diagnostic test for poor outcome [53] (Table 2).

More recently, Patil et al. measured UCH-L1 in a cohort of PA infants potentially suitable for HT. They found high UCH-L1 in PA infants with abnormal aEEG subjected to HT and normal levels in PA infants with normal aEEG [146].

Conclusions

Today, a reliable parameter able to predict perinatal brain damage in high-risk newborns is still eagerly awaited. NBs seem promising tools to provide useful information to front-line physicians in daily clinical practice. However, despite the large number of studies reported here, only a few NICUs include NBs in daily clinical practice. To date, no clinical protocols or guidelines regarding treatment in the perinatal period have been approved. Conversely, several biomarkers (S100B, GFAP, UCH-L1) have recently been approved by European and US health care agencies [149] for use with adults and children, especially in cases of traumatic brain injury [150].

The fact that biomarkers are not included among standard monitoring procedures in the perinatal period, even for the early detection of short-/long-term brain damage, warrants further consideration. To the best of our knowledge, there are several cons that need to be taken into due account. Briefly, the main limitations are:

  1. the design of the trials themselves in terms of small cohort sizes, lack of multicenter investigations, heterogeneity of the neurological complications investigated, and lack of stratification according to the severity of the main perinatal diseases (IUGR, IVH, PVL, WMI, PA-HIE);

  2. the lack of conclusive data showing that NBs could support standard monitoring parameters in the early identification of cases that do or do not require HT treatment;

  3. the different techniques for assessing NBs (i.e. ELISA, electrochemiluminescence immunoassay, HPLC) in terms of reproducibility, sensitivity, specificity and optimal timing for obtaining results, each of which may provide different results for a single patient or sample. The point is crucial for the accuracy of patients’ diagnoses and the identification of those suitable for a specific treatment at the appropriate time;

  4. the lack of consensus among manufacturers for the validation of non-invasive methods of performing assays in biological fluids such as urine and saliva that constitute the best option for less stressful longitudinal brain monitoring,

  5. the lack of reference curves for the period of investigation for each biomarker, technique and biological fluid.

The solution thus lies in multidisciplinary cooperation among neonatologists, pediatricians, biochemists, neuroscientists and manufacturers. Once such a multidisciplinary team can clarify these points, we believe it should be possible to answer the question as to what more is needed. Some authors have suggested that a panel of multiple biomarkers, rather than a single one, could provide additional accuracy in the early assessment of cases at risk for perinatal brain damage. This is especially true for PA: bearing in mind the timing of the cascade of events that leads to brain injury, the possibility that a panel of biomarkers could provide useful information in the delicate post-insult phases is more than a mere hypothesis. Studies in both animal models and humans have shown that neuro-proteins, calcium-binding proteins, vasoactive agents and oxidative stress markers are all involved, at different times, in the known phases of pathophysiological PA leading to brain damage. The value of additional biomarkers may lie in their potential to provide useful information for the validation of future therapeutic strategies. In this regard the possibility of a complex therapeutic protocol able to support neuro-protection through multiple treatments at different time-points and for different lengths of administration has been suggested for the first 96 h from an insult. Studies are in progress and the results are eagerly awaited.

We are not claiming that any one marker is of major clinical significance; we are reporting the state of the art regarding the principal biomarkers whose use in the perinatal period is discussed in recent FDA, EMA and NIH statements (Table 1). We are of course aware that there is increasing knowledge concerning new biomarkers such as Tau protein, spectrin breakdown products and inflammatory cytokines (i.e. IL 6, etc.). Despite the promising results, further investigations are needed before they can be considered to meet international health agencies’ requirements.

Table 3 shows a detailed description of biomarkers and their fulfillment of FDA and EMA criteria. The findings identify differences in laboratory performance that need further investigation. One major point regards the possibility of using biomarkers for assessment in different biological fluids. Today, S100B protein is the only biomarker that has been reported to be detectable in both urine and saliva whilst no data have been reported regarding the use of other markers in fluids, except AcA, which has been measured in urine. Another crucial issue is reproducibility, as is the time required to obtain the results. Bearing in mind the known pathophysiological steps leading to perinatal brain damage, results need to be obtained as early as possible. The ELISA and HPLC techniques can currently provide results within 2–6 h, whilst the electrochemiluminescence immunoassay offers S100B results within 2 h (median 45 min).

Table 3:

Optimality items for an NB according to the FDA and the EMA criteria: marker of brain damage and of degree and extension of the lesion; possibility of longitudinal monitoring; studied in pediatric/neonatal populations and provided of reference curves; measurable by commercial kits; measurable in different biological fluids.

NB Brain damage Degree of injury Lesion extension Longitudinal monitoring Pediatric population Available kit Reference curve Biological fluid Results output, h
S100B Y Y Y Y Y Y Ya CSF, AF, CB, PB, U, S, M <1
AM Y N N Y N Y N CSF, A, C, P <2
AcA Y N N Y N Y N CSF, A, C, P, U, M <2
NSE Y Y N Y N Y N CSF, C, P <2
OSM Y Y N N N Y N CSF, C, P <2
GFAP Y N N Y N Y N CSF, C, P <2
UCH-L1 Y Y Y Y N Y N C, P <2
  1. AM, adrenomedullin; AcA, activin A; NSE, neuron specific enolase; OSM, oxidative stress markers; G-FAP, glial fibrillary acid protein; UCH-L1, ubiquitin carboxyl-terminal hydrolase L1; Y, yes; N, no; CSF, cerebrospinal fluid; AF, amniotic fluid; CB, cord blood; NB, neurobiomarkers; PB, peripheral blood; U, urine; S, saliva; M, milk. A, Waiting for studies in wider healthy populations.

Last but not least, the costs of sampling are another relevant point: to the best of our knowledge there are no significant differences among different techniques. It is noteworthy that the cost/benefit of each biochemical marker is lower than that of any of the standard monitoring procedures currently used for monitoring the brains of sick newborns and children.

Future prospects

Recent advances in laboratory technology and performance suggest that, despite the limitations mentioned, we are not so far from reaching the target: the inclusion of biomarkers in clinical guidelines for perinatal patients in the same way that they are for adult and pediatric patients. Among new diagnostic tools metabolomics appears to offer a highly promising research field and interesting preliminary results have been reported in high-risk newborns [151]. However, further investigations are needed in wider populations before this approach can start to fulfill FDA, EMA and NIH criteria and be included in clinical guidelines.

The new challenge for the multidisciplinary team will regard the improvement of techniques for the measurement of biomarkers and the choice of biological fluid for assessment. The former will involve, at the very least: (i) identification of the sample volume required for the measurement of biomarkers, and (ii) the time required for results to be ready, in order to be able to select the cases suitable for treatment and start therapeutic strategies with the least possible delay. In addition, the possibility to measure a panel of biomarkers simultaneously, especially during different PA phases, is highly desirable.

The latter issue is an especially interesting avenue of investigation. Ideally, all biomarkers suitable for inclusion in clinical guidelines should be measurable in biological fluids that can be collected using non-invasive techniques, such as urine and saliva, in order to ensure that longitudinal monitoring of the brains of sick newborns is as accurate and useful as possible. However, bearing in mind that the principal goal of perinatologists is prevention, additional data on the assessment of S100B and other biomarkers in maternal blood in high-risk pregnancies will remain a key area of investigation aimed at determining the timing of insults and the relevant treatments as early as possible. Nonetheless, the assessment of biomarkers in maternal blood will offer physicians useful information on fetal-maternal well-being and on what is currently considered the prime objective of obstetricians: the optimal timing of delivery.

Finally, as the usefulness of biomarkers in evaluating the effectiveness/side-effects on the CNS of therapeutic strategies has been shown, it is reasonable to suggest that the longitudinal assessment of these proteins be adopted for monitoring fetal/neonatal well being in both healthy and high-risk cases [152].


Corresponding author: Diego Gazzolo, MD, PhD, Neonatal Intensive Care Unit, G. d’Annunzio University, Chieti, Italy; and Neonatal Intesive Care Unit, AO S.S. Antonio, Biagio, C. Arrigo Hospital, Spalto Marengo 46, 15100 Alessandria, Italy, Phone: +39 (131) 207241, Fax: +39 (131) 207268

Acknowledgments

This work is part of the I.O. PhD International Program under the auspices of the Italian Society of Neonatology and was partially supported by grants to DG from “I Colori della Vita Foundation”, Italy.

  1. Author contribution: Bersani Iliana contributed to conceptualization, writing – original draft. Pluchinotta Francesca contributed to conceptualization, writing – review and editing. Andrea Dotta, Savarese Immacolata, Cinzia Auriti, Natalia Chukhlantseva, Fiammetta Piersigilli, Francesca Gazzolo, contributed to data report analysis. Alessandro Varrica, Angela Satriano contributed to data investigation selection and conceptualization. Diego Gazzolo contributed to conceptualization, investigation, supervision, writing – review and editing. All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: This work is part of the I.O. PhD International Program under the auspices of the Italian Society of Neonatology and was partially supported by grants to DG from “I Colori della Vita Foundation”, Italy. We thank Diasorin, Saluggia, Italy, for supporting analysis kits.

  3. Employement or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organizations played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Gazzolo D, Abella R, Marinoni E, Di Iorio R, Li Volti G, Galvano F, et al. New markers of neonatal neurology. J Matern Fetal Neonatal Med 2009;22:57–61.10.1080/14767050903181468Search in Google Scholar PubMed

2. Serpero LD, Bellissima V, Colivicchi M, Sabatini M, Frigiola A, Ricotti A, et al. Next generation biomarkers for brain injury. J Matern Fetal Neonatal Med 2013;26:44–9.10.3109/14767058.2013.829688Search in Google Scholar PubMed

3. Mir IN, Chalak LF. Serum biomarkers to evaluate the integrity of the neurovascular unit. Early Hum Dev 2014;90:707–11.10.1016/j.earlhumdev.2014.06.010Search in Google Scholar PubMed

4. Bersani I, Auriti C, Ronchetti MP, Prencipe G, Gazzolo D, Dotta A. Use of early biomarkers in neonatal brain damage and sepsis: state of the art and future perspectives. Biomed Res Int 2015;2015:253520.10.1155/2015/253520Search in Google Scholar PubMed PubMed Central

5. Gazzolo D, Li Volti G, Gavilanes AW, Scapagnini G. Biomarkers of brain function and injury: biological and clinical significance. Biomed Res Int 2015;2015:389023.10.1155/2015/389023Search in Google Scholar PubMed PubMed Central

6. Lu H, Wang Q, Wu S, Yang L, Ren P, Yang Y, et al. Neonatal hypoxic ischemic encephalopathy-related biomarkers in serum and cerebrospinal fluid. Clin Chim Acta 2015;450:282–97.10.1016/j.cca.2015.08.021Search in Google Scholar PubMed

7. Douglas-Escobar M, Weiss MD. Biomarkers of brain injury in the premature infant. Front Neurol 2013;3:185.10.3389/fneur.2012.00185Search in Google Scholar PubMed PubMed Central

8. Douglas-Escobar M, Weiss MD. Biomarkers of hypoxic-ischemic encephalopathy in newborns. Front Neurol 2012;3:144.10.3389/fneur.2012.00144Search in Google Scholar PubMed PubMed Central

9. Graham EM, Burd I, Everett AD, Northington FJ. Blood biomarkers for evaluation of perinatal encephalopathy. Front Pharmacol 2016;7:196.10.3389/fphar.2016.00196Search in Google Scholar PubMed PubMed Central

10. Satriano A, Pluchinotta F, Gazzolo F, Serpero L, Gazzolo D. The potentials and limitations of neuro-biomarkers as predictors of outcome in neonates with birth asphyxia. Early Hum Dev 2017;105:63–7.10.1016/j.earlhumdev.2016.12.005Search in Google Scholar PubMed

11. Graham EM, Everett AD, Delpech JC, Northington FJ. Blood biomarkers for evaluation of perinatal encephalopathy: state of the art. Curr Opin Pediatr 2018;30:199–203.10.1097/MOP.0000000000000591Search in Google Scholar

12. Moore BW. A soluble protein characteristic of the nervous system. Biochem Biophys Res Commun 1965;19:739–44.10.1016/0006-291X(65)90320-7Search in Google Scholar

13. Rickmann M, Wolff JR. S100 protein expression in subpopulations of neurons of rat brain. Neuroscience 1995;67:977–91.10.1016/0306-4522(94)00615-CSearch in Google Scholar

14. Yang Q, Hamberger A, Hyden H, Wang S, Stigbrand T, Haglid KG. S-100 beta has a neuronal localisation in the rat hindbrain revealed by an antigen retrieval method. Brain Res 1995;696:49–61.10.1016/0006-8993(95)00755-FSearch in Google Scholar

15. Jonsson H, Johnsson P, Hoglund P, Alling C, Blomquist S. Elimination of S100B and renal function after cardiac surgery. J Cardiothorac Vasc Anesth 2000;14:698–701.10.1053/jcan.2000.18444Search in Google Scholar PubMed

16. Gazzolo D, Bruschettini M, Corvino V, Oliva R, Sarli R, Lituania M, et al. S100B protein concentrations in amniotic fluid correlate with gestational age and with cerebral ultrasound scanning results in healthy fetuses. Clin Chem 2001;47:954–6.10.1093/clinchem/47.5.954Search in Google Scholar

17. Aurell A, Rosengren LE, Karlsson B, Olsson JE, Zbornikova V, Haglid KG. Determination of S-100 and glial fibrillary acidic protein concentrations in cerebrospinal fluid after brain infarction. Stroke 1991;22:1254–8.10.1161/01.STR.22.10.1254Search in Google Scholar PubMed

18. Gazzolo D, Vinesi P, Marinoni E, Di Iorio R, Marras M, Lituania M, et al. S100B protein concentrations in cord blood: correlations with gestational age in term and preterm deliveries. Clin Chem 2000;46:998–1000.10.1093/clinchem/46.7.998Search in Google Scholar

19. Gazzolo D, Bruschettini M, Lituania M, Serra G, Gandullia E, Michetti F. S100B protein in urine is correlated with gestational age in healthy preterm and term newborns. Clin Chem 2001;47:1132–3.10.1093/clinchem/47.6.1132Search in Google Scholar

20. Gazzolo D, Michetti F, Bruschettini M, Marchese N, Lituania M, Mangraviti S, et al. Pediatric concentrations of S100B protein in blood:age- and sex-related changes. Clin Chem 2003;49:967–70.10.1373/49.6.967Search in Google Scholar PubMed

21. Gazzolo D, Lituania M, Bruschettini M, Ciotti S, Sacchi R, Serra G, et al. S100B protein levels in saliva: correlation with gestational age in normal term and preterm newborns. Clin Biochem 2005;38:229–33.10.1016/j.clinbiochem.2004.12.006Search in Google Scholar

22. Gazzolo D, Monego G, Corvino V, Bruschettini M, Bruschettini P, Zelano G, et al. Human milk contains S100B protein. Biochim Biophys Acta 2003;1619:209–12.10.1016/S0304-4165(02)00499-3Search in Google Scholar

23. Lu H, Huang W, Chen X, Wang Q, Zhang Q, Chang M. Relationship between premature brain injury and multiple biomarkers in cord blood and amniotic fluid. J Matern-Fetal Neonatal Med 2018;31:2898–904.10.1080/14767058.2017.1359532Search in Google Scholar

24. Haglid KG, Yang Q, Hamberger A, Bergman S, Widerberg A, Danielsen N. S-100 stimulates neurite outgrowth in the rat sciatic nerve grafted with acellular muscle transplants. Brain Res 1997;753:196–201.10.1016/S0006-8993(96)01463-1Search in Google Scholar

25. Hu J, Ferreira A, Van Eldik LJ. S100b induces neuronal death through nitric oxide release from astrocytes. J Neurochem 1997;69:2294–301.10.1046/j.1471-4159.1997.69062294.xSearch in Google Scholar PubMed

26. Michetti F, Corvino V, Geloso MC, Lattanzi W, Bernardini C, Serpero L, et al. The S100B protein in biological fluids: more than a lifelong biomarker of brain distress. J Neurochem 2012;120:644–59.10.1111/j.1471-4159.2011.07612.xSearch in Google Scholar PubMed

27. Michetti F, D’Ambrosi N, Toesca A, Puglisi MA, Serrano A, Marchese E, et al. The S100B story: from biomarker to active factor in neural injury. J Neurochem 2018;148:168–87.10.1111/jnc.14574Search in Google Scholar PubMed

28. Whitelaw A, Rosengren L, Blennow M. Brain specific proteins in posthaemorrhagic ventricular dilatation. Arch Dis Child Fetal Neonatal Ed 2001;84:F90–91.10.1136/fn.84.2.F90Search in Google Scholar

29. Whitelaw A. Intraventricular haemorrhage and posthaemorrhagic hydrocephalus: pathogenesis, prevention and future interventions. Semin Neonatol 2001;6:135–46.10.1053/siny.2001.0047Search in Google Scholar PubMed

30. Blennow M, Sävman K, Ilves P, Thoresen M, Rosengren L. Brain-specific proteins in the cerebrospinal fluid of severely asphyxiated newborn infants. Acta Paediatr 2001;90:1171–5.10.1111/j.1651-2227.2001.tb03249.xSearch in Google Scholar

31. Nagdyman N, Kömen W, Ko HK, Müller C, Obladen M. Early biochemical indicators of hypoxic-ischemic encephalopathy after birth asphyxia. Pediatr Res 2001;49:502–6.10.1203/00006450-200104000-00011Search in Google Scholar PubMed

32. Massaro AN, Chang T, Kadom N, Tsuchida T, Scafidi J, Glass P, et al. Biomarkers of brain injury in neonatal encephalopathy treated with hypothermia. J Pediatr 2012;161:434–40.10.1016/j.jpeds.2012.02.047Search in Google Scholar PubMed PubMed Central

33. Gazzolo D, Bruschettini M, Lituania M, Serra G, Bonacci W, Michetti F. Increased urinary S100B protein as an early indicator of intraventricular hemorrhage in preterm infants: correlation with the grade of hemorrhage. Clin Chem 2001;47:1836–8.10.1093/clinchem/47.10.1836Search in Google Scholar

34. Florio P, Marinoni E, Di Iorio R, Bashir M, Ciotti S, Sacchi R, et al. Urinary S100B protein concentrations are increased in intrauterine growth-retarded newborns. Pediatrics 2006;118:e747–54.10.1542/peds.2005-2875Search in Google Scholar PubMed

35. Gazzolo D, Florio P, Ciotti S, Marinoni E, di Iorio R, Bruschettini M, et al. S100B protein in urine of preterm newborns with ominous outcome. Pediatr Res 2005;58:1170–4.10.1203/01.pdr.0000185131.22985.30Search in Google Scholar PubMed

36. Gazzolo D, Marinoni E, Di Iorio R, Bruschettini M, Kornacka M, Lituania M, et al. Measurement of urinary S100B protein concentrations for the early identification of brain damage in asphyxiated full-term infants. Arch Pediatr Adolesc Med 2003;157:1163–8.10.1001/archpedi.157.12.1163Search in Google Scholar PubMed

37. Gazzolo D, Marinoni E, Di Iorio R, Bruschettini M, Kornacka M, Lituania M, et al. Urinary S100B protein measurements: a tool for the early identification of hypoxic-ischemic encephalopathy in asphyxiated full-term infants. Crit Care Med 2004;32:131–6.10.1097/01.CCM.0000104116.91462.CDSearch in Google Scholar PubMed

38. Gazzolo D, Frigiola A, Bashir M, Iskander I, Mufeed H, Aboulgar H, et al. Diagnostic accuracy of S100B urinary testing at birth in full-term asphyxiated newborns to predict neonatal death. PLoS One 2009;4:e4298.10.1371/journal.pone.0004298Search in Google Scholar PubMed PubMed Central

39. Alshweki A, Pérez-Muñuzuri A, López-Suárez O, Baña A, Couce ML. Relevance of urinary S100B protein levels as a short-term prognostic biomarker in asphyxiated infants treated with hypothermia. Medicine 2017;96:e8453.10.1097/MD.0000000000008453Search in Google Scholar PubMed PubMed Central

40. Gazzolo D, Pluchinotta F, Bashir M, Aboulgar H, Said HM, Iman I, et al. Neurological abnormalities in full-term asphyxiated newborns and salivary S100B testing: the “Cooperative Multitask against Brain Injury of Neonates” (CoMBINe) international study. PLoS One 2015;10:e0115194.10.1371/journal.pone.0115194Search in Google Scholar PubMed PubMed Central

41. Florio P, Abella R, Marinoni E, Di Iorio R, Letizia C, Meli M, et al. Adrenomedullin blood concentrations in infants subjected to cardiopulmonary bypass: correlation with monitoring parameters and prediction of poor neurological outcome. Clin Chem 2008;54:202–6.10.1373/clinchem.2007.087700Search in Google Scholar PubMed

42. Abella R, Satriano A, Frigiola A, Varrica A, Gavilanes AD, Zimmermann LJ, et al. Adrenomedullin alterations related to cardiopulmonary bypass in infants with low cardiac output syndrome. J Matern Fetal Neonatal Med 2012;25:2756–61.10.3109/14767058.2012.718393Search in Google Scholar PubMed

43. Florio P, Luisi S, Bruschettini M, Grutzfeld D, Dobrzanska A, Bruschettini P, et al. Cerebrospinal fluid activin a measurement in asphyxiated full-term newborns predicts hypoxic ischemic encephalopathy. Clin Chem 2004;50:2386–9.10.1373/clinchem.2004.035774Search in Google Scholar PubMed

44. Florio P, Perrone S, Luisi S, Vezzosi P, Longini M, Marzocchi B, et al. Increased plasma concentrations of activin a predict intraventricular hemorrhage in preterm newborns. Clin Chem 2006;52:1516–21.10.1373/clinchem.2005.065979Search in Google Scholar PubMed

45. Florio P, Frigiola A, Battista R, Abdalla Ael H, Gazzolo D, Galleri L, et al. Activin A in asphyxiated full-term newborns with hypoxic ischemic encephalopathy. Front Biosci (Elite Ed) 2010;2:36–42.10.2741/e62Search in Google Scholar PubMed

46. Florio P, Abella RF, de la Torre T, Giamberti A, Luisi S, Butera G, et al. Perioperative activin A concentrations as a predictive marker of neurologic abnormalities in children after open heart surgery. Clin Chem 2007;53:982–5.10.1373/clinchem.2006.077149Search in Google Scholar PubMed

47. Sannia A, Zimmermann LJ, Gavilanes AW, Vles HJ, Calevo MG, Florio P, et al. Elevated Activin A urine levels are predictors of intraventricular haemorrhage in preterm newborns. Acta Paediatr 2013;102:e449–54.10.1111/apa.12332Search in Google Scholar PubMed

48. Florio P, Luisi S, Moataza B, Torricelli M, Iman I, Hala M, et al. High urinary concentrations of activin A in asphyxiated full-term newborns with moderate or severe hypoxic ischemic encephalopathy. Clin Chem 2007;53:520–2.10.1373/clinchem.2005.062604Search in Google Scholar PubMed

49. Celtik C, Acunaş B, Oner N, Pala O. Neuron-specific enolase as a marker of the severity and outcome of hypoxic ischemic encephalopathy. Brain Dev 2004;26:398–402.10.1016/j.braindev.2003.12.007Search in Google Scholar PubMed

50. Buonocore G, Perrone S, Longini M, Paffetti P, Vezzosi P, Gatti MG, et al. Non protein bound iron as early predictive marker of neonatal brain damage. Brain 2003;1224–30.10.1093/brain/awg116Search in Google Scholar PubMed

51. Stewart A, Tekes A, Huisman TA, et al. Glial fibrillary acidic protein as a biomarker for periventricular white matter injury. Am J Obstet Gynecol 2013;209:27.e1–7.10.1016/j.ajog.2013.02.049Search in Google Scholar

52. Chalak LF, Sánchez PJ, Adams-Huet B, Laptook AR, Heyne RJ, Rosenfeld CR. Biomarkers for severity of neonatal hypoxic-ischemic encephalopathy and outcomes in newborns receiving hypothermia therapy. J Pediatr 2014;164:468–74.10.1016/j.jpeds.2013.10.067Search in Google Scholar

53. Massaro AN, Jeromin A, Kadom N, Vezina G, Hayes RL, Wang KK, et al. Serum biomarkers of MRI brain injury in neonatal hypoxic ischemic encephalopathy treated with whole-body hypothermia: a pilot study. Pediatr Crit Care Med 2013;14:310–7.10.1097/PCC.0b013e3182720642Search in Google Scholar

54. Jiang SH, Wang JX, Zhang YM, Jiang HF. Effect of hypothermia therapy on serum GFAP and UCH-L1 levels in neonates with hypoxic-ischemic encephalopathy. Zhongguo Dang Dai Er Ke Za Zhi 2014;16:1193–6.Search in Google Scholar

55. Douglas-Escobar MV, Heaton SC, Bennett J, Young LJ, Glushakova O, Xu X. UCH-L1 and GFAP serum levels in neonates with hypoxic-ischemic encephalopathy: a single center pilot study. Front Neurol 2014;5:273–81.10.3389/fneur.2014.00273Search in Google Scholar

56. Douglas-Escobar M, Yang C, Bennett J, Shuster J, Theriaque D, Leibovici A, et al. A pilot study of novel biomarkers in neonates with hypoxic-ischemic encephalopathy. Pediatr Res 2010;68:531–6.10.1203/PDR.0b013e3181f85a03Search in Google Scholar

57. Costantine MM, Weiner SJ, Rouse DJ, Hirtz DG, Varner MW, Spong CY, et al. Umbilical cord blood biomarkers of neurologic injury and the risk of cerebral palsy or infant death. Int J Dev Neurosci Off J Int Soc Dev Neurosci 2011;29:917–22.10.1016/j.ijdevneu.2011.06.009Search in Google Scholar

58. Gazzolo D, Vinesi P, Bartocci M, Geloso MC, Bonacci W, Serra G, et al. Elevated S100 blood level as an early indicator of intraventricular hemorrhage in preterm infants. Correlation with cerebral Doppler velocimetry. J Neurol Sci 1999;170:32–5.10.1016/S0022-510X(99)00194-XSearch in Google Scholar

59. Gazzolo D, Marinoni E, di Iorio R, Lituania M, Bruschettini PL, Michetti F. Circulating S100beta protein is increased in intrauterine growth-retarded fetuses. Pediatr Res 2002;51:215–9.10.1203/00006450-200202000-00015Search in Google Scholar PubMed

60. Giussani DA, Thakor AS, Frulio R, Gazzolo D. Acute hypoxia increases S100beta protein in association with blood flow redistribution away from peripheral circulations in fetal sheep. Pediatr Res 2005;58:179–84.10.1203/01.PDR.0000169999.66157.C0Search in Google Scholar PubMed

61. Thakor AS, Gazzolo D, Frulio R, Giussani DA. The relation of S100beta and metabolic and endocrine responses to acute fetal hypoxemia. Front Biosci (Elite Ed) 2010;2:59–67.10.2741/e66Search in Google Scholar

62. Zaigham M, Lundberg F, Olofsson P. Protein S100B in umbilical cord blood as a potential biomarker of hypoxic-ischemic encephalopathy in asphyxiated newborns. Early Hum Dev 2017;112:48–53.10.1016/j.earlhumdev.2017.07.015Search in Google Scholar PubMed

63. Summanen M, Seikku L, Rahkonen P, Stefanovic V, Teramo K, Andersson S, et al. Comparison of umbilical serum copeptin relative to erythropoietin and S100B as asphyxia biomarkers at birth. Neonatology 2017;112:60–6.10.1159/000456063Search in Google Scholar

64. Gazzolo D, Di Iorio R, Marinoni E, Masetti P, Serra G, Giovannini L, et al. S100B protein is increased in asphyxiated term infants developing intraventricular hemorrhage. Crit Care Med 2002;30:1356–60.10.1097/00003246-200206000-00037Search in Google Scholar

65. Sannia A, Risso FM, Zimmermann LJ, Gavilanes AW, Vles HJ, Gazzolo D. S100B urine concentrations in late preterm infants are gestational age and gender dependent. Clin Chim Acta 2013;417:31–4.10.1016/j.cca.2012.12.013Search in Google Scholar

66. Lee SK, Kim EC, Chi JG, Hashimura K, Mori M. Immunohistochemical detection of S-100, S-100 alpha, S-100 beta proteins, glial fibrillary acidic protein, and neuron specific enolase in the prenatal and adult human salivary glands. Pathol Res Pract 1993;189:1036–43.10.1016/S0344-0338(11)80677-1Search in Google Scholar

67. Humphrey SP, Williamson RI. A review of saliva: normal composition, flow and function. J Prosthet Dent 2001;85:162–9.10.1067/mpr.2001.113778Search in Google Scholar PubMed

68. Gazzolo D, Kornacka M, Bruschettini M, Lituania M, Giovannini L, Serra G, et al. Maternal glucocorticoid supplementation and S100B protein concentrations in cord blood and urine of preterm infants. Clin Chem 2003;49:1215–8.10.1373/49.7.1215Search in Google Scholar PubMed

69. Gazzolo D, Bruschettini M, Di Iorio R, Marinoni E, Lituania M, Marras M, et al. Maternal nitric oxide supplementation decreases cord blood S100B in intrauterine growth-retarded fetuses. Clin Chem 2002;48:647–50.10.1093/clinchem/48.4.647Search in Google Scholar

70. Bellissima V, Visser GH, Ververs TF, Bel Fv, Termote JU, Heide Mv, et al. Antenatal maternal antidepressants drugs affect S100B concentrations in fetal-maternal biological fluids. CNS Neurol Disord Drug Targets 2015;14:49–54.10.2174/1871527314666150116114033Search in Google Scholar PubMed

71. Gazzolo D, Serpero LD, Frigiola A, Abella R, Dro Giamberti A, Li Volti G, et al. Antenatal glucocorticoids supplementation and central nervous system development. Curr Drug Metab 2013;14:160–6.10.2174/1389200211314020002Search in Google Scholar

72. Sannia A, Risso FM, Serpero LD, Frulio R, Michetti F, Abella R, et al. Antenatal glucocorticoid treatment affects preterm infants’ S100B urine concentration in a dose-dependent manner. Clin Chim Acta 2010;411:1539–41.10.1016/j.cca.2010.05.045Search in Google Scholar PubMed

73. Bellissima V, Ververs TF, Visser GH, Gazzolo D. Selective serotonin reuptake inhibitors in pregnancy. Curr Med Chem 2012;19:4554–61.10.2174/092986712803306349Search in Google Scholar PubMed

74. Lattimore KA, Donn SM, Kaciroti N, Kemper AR, Neal Jr CR, Vazquez DM. Selective serotonin reuptake inhibitor (SSRI) use during pregnancy and effects on the fetus and newborn: a meta-analysis. J Perinatol 2005;25:595–604.10.1038/sj.jp.7211352Search in Google Scholar PubMed

75. Austin MP, Kildea S, Sullivan E. Maternal mortality and psychiatric morbidity in the perinatal period: challenges and opportunities for prevention in the Australian setting. Med J Aust 2007;186:364–7.10.5694/j.1326-5377.2007.tb00940.xSearch in Google Scholar PubMed

76. Austin MP. To treat or not to treat: maternal depression. SSRI use in pregnancy and adverse neonatal effects. Psychol Med 2006;36:1663–70.10.1017/S003329170600835XSearch in Google Scholar PubMed

77. Azzopardi DV, Strohm B, Edwards AD, Dyet L, Halliday HL, Juszczak E, et al. Moderate hypothermia to treat perinatal asphyxial encephalopathy. N Engl J Med 2009;361:1349–58.10.1056/NEJMoa0900854Search in Google Scholar PubMed

78. Edwards AD, Brocklehurst P, Gunn AJ, Halliday H, Juszczak E, Levene M, et al. Neurological outcomes at 18 months of age after moderate hypothermia for perinatal hypoxic ischaemic encephalopathy: synthesis and meta-analysis of trial data. Br Med J 2010;340:c363.10.1136/bmj.c363Search in Google Scholar PubMed PubMed Central

79. Massaro AN, Chang T, Baumgart S, McCarter R, Nelson KB, Glass P. Biomarkers S100B and neuron-specific enolase predict outcome in hypothermia-treated encephalopathic newborns. Pediatr Crit Care Med 2014;15:615–22.10.1097/PCC.0000000000000155Search in Google Scholar PubMed PubMed Central

80. Roka A, Kelen D, Halasz J, Beko G, Azzopardi D, Szabo M. Serum S100B and neuron-specific enolase levels in normothermic and hypothermic infants after perinatal asphyxia. Acta Paediatr 2012;101:319–23.10.1111/j.1651-2227.2011.02480.xSearch in Google Scholar PubMed

81. Massaro AN, Wu YW, Bammler TK, Comstock B, Mathur A, McKinstry RC, et al. Plasma biomarkers of brain injury in neonatal hypoxic-ischemic encephalopathy. J Pediatr 2018;194:67–75.10.1016/j.jpeds.2017.10.060Search in Google Scholar PubMed

82. Çelik Y, Atıcı A, Gülaşı S, Makharoblıdze K, Eskandari G, Sungur MA, et al. The effects of selective head cooling versus whole-body cooling on some neural and inflammatory biomarkers: a randomized controlled pilot study. Ital J Pediatr 2015;41:79.10.1186/s13052-015-0188-5Search in Google Scholar PubMed PubMed Central

83. Kitamura K, Kangawa K, Kawamoto M, Ichiki Y, Nakamura S, Matsuo H, et al. Adrenomedullin: a novel hypotensive peptide isolated from human pheochromocytoma. Biochem. Biophys Res Commun 1993;192:553.10.1006/bbrc.1993.1451Search in Google Scholar PubMed

84. Satoh F, Takahashi K, Murakami O, Totsune K, Sone M, Ohneda M, et al. Immunocytochemical localization of adrenomedullin-like immunoreactivity in the human hypothalamus and the adrenal gland. Neurosci Lett 1996;203:207–10.10.1016/0304-3940(95)12294-XSearch in Google Scholar

85. Totsune K, Takahashi K, Mackenzie HS, Murakami O, Arihara Z, Sone M, et al. Increased gene expression of adrenomedullin and adrenomedullin-receptor complexes, receptor-activity modifying protein (RAMP) 2 and calcitonin-receptor-like receptor (CRLR) in the hearts of rats with congestive heart failure. Clin Sci 2000;99:541–6.10.1042/CS20000108Search in Google Scholar

86. Nagata D, Hirata Y, Suzuki E, Kakoki M, Hayakawa H, Goto A, et al. Hypoxia-induced adrenomedullin production in the kidney. Kidney Int 1999;55:1259–67.10.1046/j.1523-1755.1999.00361.xSearch in Google Scholar

87. Allen MA, Smith PM, Ferguson AV. Adrenomedullin microinjection into the area postrema in- creases blood pressure. Am J Physiol 1997;272:1698–703.10.1152/ajpregu.1997.272.6.R1698Search in Google Scholar

88. Kitamura K, Kangawa K, Kawamoto M, Ichiki Y, Nakamura S, Matsuo H, et al. Adrenomedullin: a novel hypotensive peptide isolated from human pheochromocytoma. Biochem Biophys Res Commun 2012;425:548–55.10.1016/j.bbrc.2012.08.022Search in Google Scholar

89. Rudolph AM. Adrenomedullin: its role in perinatal adaptation. Acta Paediatr 1998;87:235–6.10.1111/j.1651-2227.1998.tb01430.xSearch in Google Scholar

90. Marinoni E, Di Iorio R, Alò P, Villaccio B, Alberini A, Cosmi EV. Immunohistochemical localization of adrenomedullin in fetal and neonatal lung. Pediatr Res 1999;45:282–5.10.1203/00006450-199902000-00021Search in Google Scholar

91. Boldt T, Luukkainen P, Fyhrquist F, Pohjavuori M, Andersson S. Birth stress increases adrenomedullin in the newborn. Acta Paediatr 1998;87:93–4.10.1111/j.1651-2227.1998.tb01393.xSearch in Google Scholar

92. Wang X, Yue TL, Barone FC, White RF, Clark RK, Willette RN, et al. Discovery of adrenomedullin in rat ischemic cortex and evidence for its role in exacerbating focal brain ischemic damage. Proc Natl Acad Sci USA 1995;92:11480–4.10.1073/pnas.92.25.11480Search in Google Scholar

93. Robertson CL, Minamino N, Ruppel RA, Kangawa K, Adelson PD, Tsuji T, et al. Increased adrenomedullin in cerebrospinal fluid after traumatic brain injury in children: a preliminary report. Acta Neurochir 2000;76:419–21.10.1007/978-3-7091-6346-7_87Search in Google Scholar

94. Kis B, Abrahám CS, Deli MA, Kobayashi H, Wada A, Niwa M, et al. Adrenomedullin in the cerebral circulation. Peptides 2001;22:1825–34.10.1016/S0196-9781(01)00533-2Search in Google Scholar

95. Di Iorio R, Marinoni E, Letizia C, Gazzolo D, Lucchini C, Cosmi EV. Adrenomedullin is increased in the fetoplacental circulation in intrauterine growth restriction with abnormal umbilical artery waveforms. Am J Obstet Gynecol 2000;182:650–4.10.1067/mob.2000.103944Search in Google Scholar

96. Gazzolo D, Marinoni E, Giovannini L, Letizia C, Serra G, Di Iorio R, et al. Circulating adrenomedullin is increased in preterm newborns developing intraventricular hemorrhage. Pediatr Res 2001;50:544–7.10.1203/00006450-200110000-00020Search in Google Scholar

97. Kamata S, Kamiyama M, Usui N, Kitayama Y, Okuyama H, Kubota A, et al. Is adrenomedullin involved in the pathophysiology of persistent pulmonary hypertension of the newborn? Pediatr Surg Int 2004;20:24–6.10.1007/s00383-003-1071-9Search in Google Scholar

98. Di Iorio R, Marinoni E, Lituania M, Serra G, Letizia C, Cosmi EV, et al. Adrenomedullin increases in term asphyxiated newborns developing intraventricular hemorrhage. Clin Biochem 2004;37:1112–6.10.1016/j.clinbiochem.2004.04.011Search in Google Scholar

99. Erdinc K, Sarici SU, Akgul EO, Agilli M, Ozcan O. Relationship between neonatal adrenomedullin and bilirubin levels. J Matern Fetal Neonatal Med 2014;27:30–5.10.3109/14767058.2013.799655Search in Google Scholar

100. Luisi S, Florio P, Reis FM, Petraglia F. Expression and secretion of activin A: possible physiological and clinical implications. Eur J Endocrinol 2001;145:225–36.10.1530/eje.0.1450225Search in Google Scholar

101. Luisi S, Calonaci G, Florio P, Lombardi I, De Felice C, Bagnoli F, et al. Identification of activin A and follistatin in human milk. Growth Factors 2002;20:147–50.10.1080/0897719021000042334Search in Google Scholar

102. Lai M, Sirimanne E, Williams CE, Gluckman PD. Sequential patterns of inhibin subunit gene expression following hypoxic ischemic injury in the rat brain. Neurosci 1996;70:1013–24.10.1016/0306-4522(95)00413-0Search in Google Scholar

103. Florio P, Gazzolo D, Luisi S, Petraglia F. Activin A in brain injury. Adv Clin Chem 2007;43:117–30.10.1016/S0065-2423(06)43004-3Search in Google Scholar

104. Schubert D, Kimura H, LaCorbiere M, Vaughan J, Karr D, Fischer WH. Activin is a nerve cell survival molecule. Nature 1990;344:868–70.10.1038/344868a0Search in Google Scholar PubMed

105. Krieglstein K, Suter-Crazzolara C, Fischer WH, Unsicker K. TGF-beta superfamily members promote survival of midbrain dopaminergic neurons and protect them against MPP+ toxicity. EMBO J 1995;14:736–42.10.1002/j.1460-2075.1995.tb07052.xSearch in Google Scholar

106. Iwahori Y, Saito H, Torii K, Nishiyama N. Activin exerts a neurotrophic effect on cultured hippocampal neurons. Brain Res 1997;760:52–8.10.1016/S0006-8993(97)00275-8Search in Google Scholar

107. Hughes PE, Alexi T, Williams CE, Clark RG, Gluckman PD. Administration of recombinant human Activin-A has powerful neurotrophic effects on select striatal phenotypes in the quinolinic acid lesion model of Huntington’s disease. Neurosci 1999;92:197–209.10.1016/S0306-4522(98)00724-6Search in Google Scholar

108. Florio P, Perrone S, Luisi S, Longini M, Tanganelli D, Petraglia F, et al. Activin a plasma levels at birth: an index of fetal hypoxia in preterm newborn. Pediatr Res 2003;54:696–700.10.1203/01.PDR.0000086905.71963.1DSearch in Google Scholar

109. Florio P, Reis FM, Severi FM, Luisi S, Imperatore A, Palumbo MA, et al. Umbilical cord serum activin A levels are increased in pre-eclampsia with impaired blood flow in the uteroplacental and fetal circulation. Placenta 2006;27:432–7.10.1016/j.placenta.2005.04.008Search in Google Scholar

110. Fiala M, Baumert M, Surmiak P, Walencka Z, Sodowska P. Umbilical activin A concentration as an early marker of perinatal hypoxia. J Matern Fetal Neonatal Med 2012;25:2098–101.10.3109/14767058.2012.675373Search in Google Scholar

111. Bellissima V, Visser GH, Ververs TF, Van Bel F, Termote JU, Van Der Heide M, et al. Antenatal maternal antidepressants drugs affect Activin A concentrations in maternal blood, in amniotic fluid and in fetal cord blood. J Matern Fetal Neonatal Med 2011;24:31–4.10.3109/14767058.2011.604931Search in Google Scholar

112. Påhlman S, Esscher T, Bergvall P, Odelstad L. Purification and characterization of human neuron-specific enolase: radioimmunoassay development. Tumour Biol 1984;5:127–39.Search in Google Scholar

113. Kaiser E, Kuzmits R, Pregant P, Burghuber O, Worofka W. Clinical biochemistry of neuron specific enolase. Clin Chim Acta 1989;183:13–31.10.1016/0009-8981(89)90268-4Search in Google Scholar

114. Berger RP, Adelson PD, Pierce MC, Dulani T, Cassidy LD, Kochanek PM. Serum neuron-specific enolase, S100B, and myelin basic protein concentrations after inflicted and noninflicted traumatic brain injury in children. J Neurosurg 2005;103:61–8.10.3171/ped.2005.103.1.0061Search in Google Scholar PubMed

115. Vos PE, Lamers KJ, Hendriks JC, Van Haaren M, Beems T, Zimmerman C, et al. Glial and neuronal proteins in serum predict outcome after severe traumatic brain injury. Neurology 2004;62:1303–10.10.1212/01.WNL.0000120550.00643.DCSearch in Google Scholar

116. Wilkinson AA, Dennis M, Simic N, Taylor MJ, Morgan BR, Frndova H, et al. Brain biomarkers and pre-injury cognition are associated with long-term cognitive outcome in children with traumatic brain injury. BMC Pediatr 2017;17:173.10.1186/s12887-017-0925-6Search in Google Scholar PubMed PubMed Central

117. Thornberg B, Thiringer K, Hagberg H, Kjellmer I. Neuron specific enolase in asphyxiated newborns: association with encephalopathy and cerebral function monitor trace. Arch Dis Child Fetal Neonatal Ed 1995;72:F39–42.10.1136/fn.72.1.F39Search in Google Scholar PubMed PubMed Central

118. Ezgü FS, Atalay Y, Gücüyener K, Tunç S, Koç E, Ergenekon E, et al. Neuron-specific enolase levels and neuroimaging in asphyxiated term newborns. J Child Neurol 2002;17:824–9.10.1177/08830738020170111301Search in Google Scholar PubMed

119. Garcia-Alix A, Cabañas F, Pellicer A, Hernanz A, Stiris TA, Quero J. Neuron-specific enolase and myelin basic protein: relationship of cerebrospinal fluid concentrations to the neurologic condition of asphyxiated full-term infants. Pediatrics 1994;93:234–40.10.1542/peds.93.2.234Search in Google Scholar

120. Trakas E, Domnina Y, Panigrahy A, Baust T, Callahan PM, Morell VO, et al. Serum neuronal biomarkers in neonates with congenital heart disease undergoing cardiac surgery. Pediatr Neurol 2017;72:56–61.10.1016/j.pediatrneurol.2017.04.011Search in Google Scholar PubMed

121. Zhang XH, Zhang BL, Guo SM, Wang P, Yang JW. Clinical significance of dynamic measurements of seric TNF-α, HMGBl, and NSE levels and aEEG monitoring in neonatal asphyxia. Eur Rev Med Pharmacol Sci 2017;21:4333–39.Search in Google Scholar

122. Perrone S, Santacroce A, Longini M, Proietti F, Bazzini F, Buonocore G. The Free Radical Diseases of Prematurity: From Cellular Mechanisms to Bedside. Oxid Med Cell Longev 2018;2018:7483062.10.1155/2018/7483062Search in Google Scholar PubMed PubMed Central

123. Buonocore G, Groenendaal F. Anti-oxidant strategies. Seminars in Fetal Neon Med 2007;12:287–95.10.1016/j.siny.2007.01.020Search in Google Scholar PubMed

124. Inder T, Mocatta T, Darlow B, Spencer C, Volpe JJ, Winterbourn C. Elevated free radical products in the cerebrospinal fluid of VLBW infants with cerebral white matter injury. Pediatr Res 2002;52:213–8.10.1203/00006450-200208000-00013Search in Google Scholar PubMed

125. Inder T, Mocatta T, Darlow B, Spencer C, Senthilmohan R, Winterbourn CC, et al. Markers of oxidative injury in the cerebrospinal fluid of a premature infant with meningitis and periventricular leukomalacia. J Pediatr 2002;140:617–21.10.1067/mpd.2002.123627Search in Google Scholar PubMed

126. Gücüyener K, Ergenekon E, Demiryürek T, Erbaş D, Oztürk G, Koç E, et al. Cerebrospinal fluid levels of nitric oxide and nitrotyrosine in neonates with mild hypoxic-ischemic encephalopathy. J Child Neurol 2002;17:815–8.10.1177/08830738020170111101Search in Google Scholar PubMed

127. Gathwala G, Marwah A, Gahlaut V, Marwah P. Effect of high-dose phenobarbital on oxidative stress in perinatal asphyxia: an open label randomized controlled trial. Indian Pediatr 2011;48:613–7.10.1007/s13312-011-0106-xSearch in Google Scholar PubMed

128. Aridas JDS, Yawno T, Sutherland AE, Nitsos I, Ditchfield M, Wong FY, et al. Systemic and transdermal melatonin administration prevents neuropathology in response to perinatal asphyxia in newborn lambs. J Pineal Res 2018;64:e12479.10.1111/jpi.12479Search in Google Scholar PubMed PubMed Central

129. Comporti M, Signorini C, Leoncini S, Buonocore G, Rossi V, Ciccoli L. Plasma F2-isoprostanes are elevated in newborns and inversely correlated to gestational age. Free Radic Biol Med 2004;37:724–32.10.1016/j.freeradbiomed.2004.06.007Search in Google Scholar PubMed

130. Signorini C, Perrone S, Sgherri C, Ciccoli L, Buonocore G, Leoncini S. Plasma esterified F2-isoprostanes and oxidative stress in newborns: role of nonprotein-bound iron. Pediatr Res 2008;63:287–91.10.1203/PDR.0b013e318163a1fdSearch in Google Scholar PubMed

131. Ferencz Á, Orvos H, Hermesz E. Major differences in the levels of redox status and antioxidant defence markers in the erythrocytes of pre- and full-term neonates with intrauterine growth restriction. Reprod Toxicol 2015;53:10–4.10.1016/j.reprotox.2015.02.008Search in Google Scholar PubMed

132. Gupta P, Narang M, Banerjee BD, Basu S. Oxidative stress in term small for gestational age neonates born to undernourished mothers: a case control study. BMC Pediatr 2004;4:14.10.1186/1471-2431-4-14Search in Google Scholar PubMed PubMed Central

133. Dede H, Takmaz O, Ozbasli E, Dede S, Gungor M. Higher level of oxidative stress markers in small for gestational age newborns delivered by cesarean section at term. Fetal Pediatr Pathol 2017;36:232–9.10.1080/15513815.2017.1303860Search in Google Scholar PubMed

134. Mondal N, Bhat BV, Banupriya C, Koner BC. Oxidative stress in perinatal asphyxia in relation to outcome. Indian J Pediatr 2010;77:515–7.10.1007/s12098-010-0059-4Search in Google Scholar PubMed

135. Perrone S, Szabó M, Bellieni CV, Longini M, Bangó M, Kelen D, et al. Whole body hypothermia and oxidative stress in babies with hypoxic-ischemic brain injury. Pediatr Neurol 2010;43:236–40.10.1016/j.pediatrneurol.2010.05.009Search in Google Scholar PubMed

136. Abdel Ghany EA, Alsharany W, Ali AA, Youness ER, Hussein JS. Anti-oxidant profiles and markers of oxidative stress in preterm neonates. Paediatr Int Child Health 2016;36:134–40.10.1179/2046905515Y.0000000017Search in Google Scholar PubMed

137. Perrone S, Mussap M, Longini M, Fanos V, Bellieni CV, Proietti F, et al. Oxidative kidney damage in preterm newborns during perinatal period. Clin Biochem 2007;40:656–60.10.1016/j.clinbiochem.2007.01.012Search in Google Scholar

138. Tsukahara H, Toyo-Oka M, Kanaya Y, Ogura K, Kawatani M, Hata A, et al. Quantitation of glutathione S transferase-pi in the urine of preterm neonates. Pediatr Int 2005;47:528–31.10.1111/j.1442-200x.2005.02123.xSearch in Google Scholar

139. Bignami A, Eng LF, Dahl D, Uyeda CT. Localization of the glial fibrillary acidic protein in astrocytes by immunofluorescence. Brain Res 1972;43:429–35.10.1016/0006-8993(72)90398-8Search in Google Scholar

140. Eng LF, Ghirnikar RS, Lee YL. Glial fibrillary acidic protein: GFAP-thirty-one years (1969–2000). Neurochem Res 2000;25:1439–51.10.1023/A:1007677003387Search in Google Scholar

141. Blennow M, Rosengren L, Jonsson S, Forssberg H, Katz-Salamon M, Hagberg H, et al. Glial fibrillary acidic protein is increased in the cerebrospinal fluid of preterm infants with abnormal neurological findings. Acta Paediatr 1996;85:485–9.10.1111/j.1651-2227.1996.tb14068.xSearch in Google Scholar PubMed

142. McKenney SL, Mansouri FF, Everett AD, Graham EM, Burd I, Sekar P. Glial fibrillary acidic protein as a biomarker for brain injury in neonatal CHD. Cardiol Young 2016;26:1282–9.10.1017/S1047951115002346Search in Google Scholar PubMed PubMed Central

143. Zaigham M, Hayes R, Undén J, Olofsson P. Umbilical cord blood concentrations of ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) and glial fibrillary acidic protein (GFAP) in neonates developing hypoxic-ischemic encephalopathy. J Matern Fetal Neonatal Med 2016;29:1822–8.10.3109/14767058.2015.1064108Search in Google Scholar PubMed

144. Looney A-M, Ahearne C, Boylan GB, Murray DM. Glial fibrillary acidic protein is not an early marker of injury in perinatal asphyxia and hypoxic-ischemic encephalopathy. Front Neurol 2015;6:26410.3389/fneur.2015.00264Search in Google Scholar PubMed PubMed Central

145. Ennen CS, Huisman TA, Savage WJ, Northington FJ, Jennings JM, Everett AD, et al. Glial fibrillary acidic protein as a biomarker for neonatal hypoxic-ischemic encephalopathy treated with whole-body cooling. Am J Obstet Gynecol 2011;205:251.10.1016/j.ajog.2011.06.025Search in Google Scholar PubMed PubMed Central

146. Patil UP, Mally PV, Wachtel EV. Serum biomarkers of neuronal injury in newborns evaluated for selective head cooling: a comparative pilot study. J Perinat Med 2018;46:942–7.10.1515/jpm-2017-0354Search in Google Scholar PubMed

147. Lewis SB, Wolper R, Chi YY, Miralia L, Wang Y, Yang C, et al. Identification and preliminary characterization of ubiquitin C terminal hydrolase 1 (UCHL1) as a biomarker of neuronal loss in aneurysmal subarachnoid hemorrhage. J Neurosci Res 2010;88:1475–84.10.1002/jnr.22323Search in Google Scholar PubMed

148. Welch RD, Ayaz SI, Lewis LM, Unden J, Chen JY, Mika VH, et al. Ability of serum glial fibrillary acidic protein, ubiquitin C-terminal hydrolase-L1, and S100B to differentiate normal and abnormal head computed tomography findings in patients with suspected mild or moderate traumatic brain injury. J Neurotrauma 2016;33:203–14.10.1089/neu.2015.4149Search in Google Scholar PubMed PubMed Central

149. Berger RP, Hayes RL, Richichi R, Beers SR, Wang KK. Serum concentrations of ubiquitin C-terminal hydrolase-L1 and αII-spectrin breakdown product 145 kDa correlate with outcome after pediatric TBI. Neurotrauma 2012;29:162–7.10.1089/neu.2011.1989Search in Google Scholar PubMed PubMed Central

150. Undén L, Calcagnile O, Undén J, Reinstrup P, Bazarian J. Validation of the Scandinavian guidelines for initial management of minimal, mild and moderate traumatic brain injury in adults. BMC Med 2015;13:292.10.1186/s12916-015-0533-ySearch in Google Scholar PubMed PubMed Central

151. Noto A, Fanos V, Dessì A. Metabolomics in newborns. Adv Clin Chem 2016;74:35–61.10.1016/bs.acc.2015.12.006Search in Google Scholar PubMed

152. Gazzolo D, Marinoni E, Di Iorio R, Lituania M, Marras M, Bruschettini M, et al. High maternal blood S100B concentrations in pregnancies complicated by intrauterine growth restriction and intraventricular hemorrhage. Clin Chem 2006;52:819–26.10.1373/clinchem.2005.060665Search in Google Scholar PubMed

Received: 2019-07-18
Accepted: 2019-10-31
Published Online: 2019-12-19
Published in Print: 2020-03-26

©2020 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 25.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2019-0725/html
Scroll to top button