Next Article in Journal
Ferroptosis Activation Contributes to the Formation of Skin Lesions in Psoriasis Vulgaris
Next Article in Special Issue
Hyperoxia Disrupts Lung Lymphatic Homeostasis in Neonatal Mice
Previous Article in Journal
Aflatoxin B1 Induced Oxidative Stress and Gut Microbiota Disorder to Increase the Infection of Cyprinid Herpesvirus 2 in Gibel Carp (Carassius auratus gibelio)
Previous Article in Special Issue
Gestational Hypertension and High-Density Lipoprotein Function: An Explorative Study in Overweight/Obese Women of the DALI Cohort
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Brain Damage in Preterm and Full-Term Neonates: Serum Biomarkers for the Early Diagnosis and Intervention

by
Serafina Perrone
1,*,
Federica Grassi
2,
Chiara Caporilli
2,
Giovanni Boscarino
2,
Giulia Carbone
2,
Chiara Petrolini
1,
Lucia Maria Gambini
1,
Antonio Di Peri
1,
Sabrina Moretti
1,
Giuseppe Buonocore
3 and
Susanna Maria Roberta Esposito
2
1
Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
2
Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
3
Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(2), 309; https://doi.org/10.3390/antiox12020309
Submission received: 13 October 2022 / Revised: 14 January 2023 / Accepted: 26 January 2023 / Published: 29 January 2023
(This article belongs to the Special Issue Oxidative Stress in Fetuses and Newborns)

Abstract

:
The Brain is vulnerable to numerous insults that can act in the pre-, peri-, and post-natal period. There is growing evidence that demonstrate how oxidative stress (OS) could represent the final common pathway of all these insults. Fetuses and newborns are particularly vulnerable to OS due to their inability to active the antioxidant defenses. Specific molecules involved in OS could be measured in biologic fluids as early biomarkers of neonatal brain injury with an essential role in neuroprotection. Although S-100B seems to be the most studied biomarker, its use in clinical practice is limited by the complexity of brain damage etiopathogenesis and the time of blood sampling in relation to the brain injury. Reliable early specific serum markers are currently lacking in clinical practice. It is essential to determine if there are specific biomarkers that can help caregivers to monitor the progression of the disease in order to active an early neuroprotective strategy. We aimed to describe, in an educational review, the actual evidence on serum biomarkers for the early identification of newborns at a high risk of neurological diseases. To move the biomarkers from the bench to the bedside, the assays must be not only be of a high sensitivity but suitable for the very rapid processing and return of the results for the clinical practice to act on. For the best prognosis, more studies should focus on the association of these biomarkers to the type and severity of perinatal brain damage.

1. Introduction

Multiple factors acting in the pre- or post-natal period (i.e., metabolic complications, nutritional intake, toxic, and infectious disease) could influence the pathophysiology of cerebral damage [1]. Many studies have been conducted to explain the fetal-neonatal perturbations that predispose to the genesis of a neurological delay or other diseases in adult life [2]. Maternal diabetes, prenatal hypoxic/ischemic events, inadequate nutritional intake in early life, early metabolic complications, inflammatory/infective insults, and transfusions are all specific triggers for the activation of oxidative stress (OS), with a rapid increase in the generation of free radicals (FRs). Therefore, these factors could be triggered in a common pathway, leading to the activation of the OS mechanism, with the final increase in FRs due to the imbalance in the antioxidant systems’ homeostasis [3]. This imbalance occurs at birth in all newborns as a consequence of the hyperoxic challenge due to the transition from the hypoxic intrauterine environment to extrauterine life [4]. The neonatal brain, especially the preterm neonatal brain, is particularly vulnerable to the insult of OS and sensitive to the dangerous effects of FRs due to the inability to activate the antioxidant defenses [5]. The study and validation of specific and optimal biomarkers of neonatal brain injury, measured in biologic fluids, is an essential step in neuroprotection. These biomarkers could improve the early identification of a newborn at a high risk of brain injury, promoting specific preventive or therapeutic treatment, as the new, modernized standard of care for high-risk neonates [6]. Serum biomarkers could help the clinician to assess the entity of the neonatal brain injury and to monitor the progression of the disease in addition to the standard procedures such as cerebral ultrasound (cUS) and magnetic resonance imaging (MRI). The aim of this educational review is to describe the actual available evidence on serum biomarkers for the early diagnosis of brain damage, analyzing also the causes of neurological diseases for preterm and full-term newborns.

2. Brain Development

The second half of gestation and the first 28 days of life are the most important developmental periods for the development of brain structures and cerebral pathways [7]. The neuropathology of brain damage evolves around multiple events that occur at 24–40 weeks of gestation, during the critical stage of cerebral maturation, when cells replication and differentiation are widely activated [8]. Cerebral morphogenesis could be schematically described in 3 steps: (1) histogenesis of the cortex and development of commissures (from 7 to 23 weeks of gestation); (2), cortical morphogenesis and white matter development (from 20 to 40 weeks of gestation); and (3) neuronal myelination (from 18 weeks of gestation to first adult life) [9,10]. Cerebral morphogenesis is finally characterized by an increase in the brain volume, by the development of sulcation, the modification of the ventricular shape, and the reduction in subarachnoid spaces. These processes can be evaluated during the fetal period with the help of fetal brain MRI [8,9]. During this process of maturation, there is also an important neuronal differentiation with synapse formation, the maturation of myelin due to glial cells differentiation, new neurotransmitters synthesis, and vascular development [7,10].
Considering the very complex characteristics of these developing phases, endogenous insults, such as ischemia, inflammation, excitotoxicity, and FRs attack, could affect the normal morphogenesis of brain structures, causing important long-term damages [8]. In addition, it has been largely demonstrated that, in the early phases of life, other events such as under-nutrition, metabolic side effects, drug exposure, and other factors typical of the neonatal intensive care could affects the post-natal cerebral maturation [11,12,13,14,15,16,17]. Considering these dynamic phases of pre-natal and post-natal brain maturation, neonatal life (especially for babies born preterm) is a critical window for long-term neurodevelopment.

3. Brain Damage in Preterm Newborn

Preterm birth interrupts the process of cerebral maturation, causing an impaired brain development and increasing the risk for neurodevelopmental delay in postnatal life.
The intraventricular hemorrhage (IVH), produced by an injury of the germinal matrix and the subventricular zone, is one of the leading causes of brain damage in preterm newborns [8]. Intraventricular hemorrhage affects 20–25% of preterm newborns with a birth weight less than 1500 gr [8]. The immaturity of blood vessels in the germinal matrix, a highly vascular cerebral region, combined with poor tissue vascular support, makes this region particularly vulnerable to the development of IVH in preterm newborns [8,18]. In full-term infants, the preferential site of IVH is instead the choroid plexus [7].
Intraventricular hemorrhage could be classified into three grades according to Volpe (Figure 1) [7]:
  • Grade 1: hemorrhage apparently confined to germinal matrix, distended less than 10% of ventricular space;
  • Grade 2: hemorrhage clearly localized in the ventricle but occupying 10–50% of ventricular space;
  • Grade 3: large hemorrhage, occupying greater than 50% of ventricle with or without ventricular echo-densities.
In addition, when the hemorrhage also includes the parenchymal tissue, the lesion (periventricular hemorrhagic infarction-PVHI) is considered separately because these abnormalities generally are not related simply by the extension of a matrix hemorrhage or IVH into the normal brain parenchyma (Figure 1) [7].
Neonates with IVH could have different clinical presentations that range from an acute deterioration with apnea, pallor, acidosis, hypotension, bulging fontanel, seizures, and decreased muscle tone to a “clinically silent syndrome” when no symptoms are presented [7].
Periventricular leukomalacia (PVL) is another important brain lesion that affects preterm newborns, causing cerebral palsy (CP), cognitive, behavioral, and attention deficits [7]. It is a cerebral white matter injury (WMI) associated with a decreased cortex volume, thalamus, and basal ganglia volume [8]. The lesion starts as a focal lesion that occurs in deep periventricular white matter zones due to neuronal necrosis, reactive gliosis, and microglial activation [7]. Necrosis can be macroscopic (several millimeters or more) and can evolve rapidly in some weeks to multiple cystic lesions, also assessable through a cerebral ultrasound scan (cUS) known as “cystic PVL” [8]. Most commonly, the focal necrosis is microscopic and evolves in several weeks to glial scars which are not visible at the beginning by the neuroimaging [7].
Neuroimaging techniques, such as cUS and MRI, are useful tools for the diagnosis of IVH and PVL [19,20]. However, both present some limitations: cUS is an operator dependent technique, while MRI is not easy to perform during the first weeks of life for the critical clinical condition of the babies and the need for a proper sedation [7,19,20]. Considering the neuroimaging limits and the possible lack of symptoms in the early stage of PVL, it would be useful to identify biomarkers capable of predicting which neonates may suffer from IVH or PVL [7,8]. Despite some studies evaluating the role of innovative neuroprotective therapy (i.e., stem cells, erythropoietin (EPO), vitamin E, and melatonin), the early diagnosis of IVH/PVL is still the gold-standard to reduce a long-term neurological consequence, such as cerebral palsy (CP) [21,22,23].
Cerebral palsy has been defined as “a group of permanent disorders of the development of movement and posture, causing activity limitation, that are attributed to nonprogressive disturbances that occurred in the developing fetal or infant brain” [24]. The prevalence of CP in preterm infants born between 22 and 31 weeks’ gestational age is of 62.5/1000 live births [25]. Despite the fact that a preterm birth is the most important risk factor for CP, the biologic basis seem to be multifactorial [26,27]. There are several perinatal interventions that aim to prevent CP, such as the administration of magnesium sulfate to women at risk of a preterm birth and methylxanthine therapy for apnea of prematurity, essential to reduce the risk of hypoxemia and bradycardia [28,29]. The diagnosis of CP can be made in the first 6 months of age and is based on clinical presentation, in association with other diagnostic tools such as neuroimaging (cUS and MRI) and early standardized motor/neurologic assessments (i.e., Hammersmith Infant Neurological Examinations) [27,30]. Despite the advance in technology in the last decade, the incidence of CP remains higher for the preterm population with a late diagnosis for the efficacy of a neuroprotective intervention. Thus, a rapid screening test, such as serum biomarkers, could be decisive to guarantee an early diagnosis in order to improve the neurological outcome of this higher risk population.

4. Brain Damage in Full-Term Newborn

The principal causes of brain damage in full-term newborns are the hypoxic ischemic encephalopathy (HIE), the neonatal stroke, and systemic infections [31]. In the pathogenesis of HIE, the timing of the injury and treatment have fundamental roles [32]. Although the introduction of therapeutic hypothermia as the gold-standard for the treatment of HIE has been an essential change in the prognosis [33,34], this approach can be started only for full-term newborns in the first 6 h of life, and it has only a partial effectiveness because 45% of patients still die or have a neurodevelopmental disability despite this treatment [35]. The first stage of HIE is characterized by a primary energy failure due to hypoxic ischemia (HI), which causes a primary energy failure. This event takes to a decrease in ATP, with an increase in the lactate levels and systemic acidosis [36,37]. Subsequently, the reperfusion of the ischemic zone determines an oxidative metabolism with the activation of inflammation and apoptotic cascades [38]. These events lead to an activation of lipases, proteases, and endonucleases. In particular, lipases induce a production of free fatty acids, especially arachidonic acid, with a release by proteases of superoxide FRs through the activation of cyclooxygenase and the production of prostaglandin [39,40]. This excessive production of FRs during the ischemia/reperfusion process leads to the further production of FRs and other toxic metabolites in larger quantities than the antioxidant capacity of cells. Finally, the third stage after an hypoxic insult, which lasts for months to years, is characterized by reactive gliosis with a persistent inflammation and epigenetic changes [21].
Another cause of near-term brain damage is stroke, involving both arterial and venous cerebral blood vessels, with a hemorrhagic, ischemic, or mixed component [41,42]. The event is symptomatic for 50% of the cases [43]. The most frequent stroke is the arterial ischemic one, but it can be followed by cerebral sinovenous thrombosis and neonatal hemorrhagic stroke [43]. The main clinical manifestation in the first weeks of life are seizures [44,45]. Despite the vulnerability of the zone near to the stroke, endogenous mechanisms, which are still not fully understood, could potentially preserve it [46]. Long term consequences are represented by seizures, CP, congenital hemiplegia, neurodevelopmental delay including intellectual disability, language retardation, and behavioral problems from infancy to adult life [47,48].
Other important causes of brain damage in the neonatal period for full-term newborns, are congenital infections, able to determine a neuronal stem cells injury during the vulnerable phase of brain development. The risk of the transmission of intrauterine during pregnancy is higher with those of an older gestational age, while the risk of adverse fetal consequences significantly increased with the transmission of infection during the first half of pregnancy [49,50]. Considering the TORCH group infections, the congenital cytomegalovirus (cCMV) infection is the primary cause of brain damage in newborns. The incidence of cCMV is higher in low–middle income countries. In addition to neurodevelopmental delay, cCMV is the principal cause of nongenetic sensorial hearing loss [51]. At birth, about 10–15% of congenitally infected infants are symptomatic and approximately half of them will present long-term consequences [52,53]. However, also the 10–15% of asymptomatic infants infected by cCMV could develop long-term sequelae [54]. There is no evidence for the effective treatment of asymptomatic infants and the use of antiviral therapy is contraindicated by an animal model for its toxicity (i.e., neutropenia, thrombocytopenia, gonadal toxicity, and carcinogenicity) [55]. Thus, it is important to find an early biomarker which is able to predict the long-term consequences to justify antiviral therapy in these populations in order to reduce the hearing loss and neurological disability related to CMV infection.

5. Serum Biomarkers

There are many serum biomarkers studied in relation to brain damage for newborns (Figure 2, Table 1):

5.1. Dipeptidylpeptidase 4

Dipeptidylpeptidase 4 (DPP4) is a membrane-bound serine protease that is localized in the gastrointestinal tract, liver, lungs, kidneys, and also on T-lymphocytes where the enzyme is known as activation marker CD26. DPP4 and DPP4-like peptidases from the prolyl oligopepetidase family hydrolyze neuropeptides, cytokines, and peptide hormones [80,81,82]. It is supposed that these enzymes could be associated with the neurodegenerative processes related to cerebral ischemia. The participation of DPP4 in the processes of inflammation and neurodegeneration in ischemic brain damage was demonstrated in animals [83].
Yakovleva et al. investigated the serum DPP4 activity in neonates with cerebral ischemia. The serum DPP4 activity in the group of patients with cerebral ischemia was significantly higher than the control group. In addition, preterm and full-term neonates show no differences in DPP4 activity under the influence of hypoxia [56].
Considering the serum DPP4 activity in neonates with different neurological symptomatology: among the full-term neonates with cerebral ischemia, the maximum DPP4 activity was found in the group with an excitement syndrome. These values were significantly higher than those in children with a depression syndrome. Instead, for the preterm infants with cerebral ischemia, there were no statistically significant differences in the serum DPP4 activity between the groups with different functional nervous system activity [56].
It might be supposed that DPP4 takes part in regulating the central nervous system activity in full-term newborns. On the contrary, in preterm newborns, these regulatory mechanisms are still in a developing stage. It is known that DPP4 takes part in the transduction of the signal for the activation of T-helper cells, which leads to their proliferation and cytokine production [84]. Thus, hypoxia could trigger an inflammatory response determining an increase in DPP4 activity in the blood [85,86].
Yakovleva et al. opens new opportunities for the development of DPP4 inhibitors for the prevention of detrimental neurological consequences in newborns with cerebral ischemia [56].

5.2. Cytokines

Hypoxic-ischemic injury activates many inflammatory processes in the brain. A number of inflammatory molecules have been suggested to be sentinel biomarkers of HIE. The serum proteins are readily measurable and may be useful biomarkers of injury phases [87].
Microglia are the resident immune cells of the brain, able to start the inflammatory response in the central nervous system [88]. HIE induces microglia cells to produce proinflammatory cytokines, with a damage to the overall structure of the brain [89]. Microglia generate not only an excess of inflammatory cytokines (e.g., TNF-a and IL-1b) but also glutamate, nitric oxide (NO), and reactive oxygen species (ROS), which collectively cause oligodendrocyte death, axonal degeneration, and the disruption of the immature blood–brain barrier [90]. In addition, many inflammatory cytokines have direct toxic effects due to the increased production of inducible NO synthase and cyclooxygenase and the release of FRs [57]. Proinflammatory cytokines might damage developing white matter by inducing intravascular coagulation and/or thrombosis and vasoconstriction or by inducing the production of other cytokines such as the platelet-activating factor [91].
In 2003, Chiesa et al. reported that IL-6 was a good marker of HIE and long-term neurodevelopmental delay [58]. Many years later, Chaparro’s study supported those of Chiesa, showing higher levels of IL-6 in hypoxic patients at birth [57,58]. Moreover, Chaparro et al. demonstrated that the expression of IL-1b and TNF-a was markedly increased by several fold in patients affected by HIE compared with healthy controls, and the IL-6 expression was significantly increased by nine-fold [57].
A Greek study, in agreement with more recent studies, showed that the asphyxiated neonates had significantly higher IL-6 and IL-1ß serum levels than healthy controls. On the contrary, the TNF-a serum levels did not differ between the two groups of neonates studied (asphyxiated neonates and healthy controls) [59]. No differences in the serum levels of TNF-a were found between the neonates who showed neurologic abnormalities and those without neurodevelopmental delay [59].

5.3. Neuron-Specific Enolase

Neuron-specific enolase (NSE) is a highly specific glycolysis isoenzyme for neurons and peripheral neuroendocrine cells [92].
The NSE level was measured as a marker of neuronal death. Clinical studies showed that the NSE concentrations are significantly increased in HIE and asphyxiated neonates compared with the healthy controls [57,61,62,63]. A significant rise in NSE in term infants with HIE seems to be associated with conspicuous neurological damage or death. Chaparro et al. revealed that the NSE level in the blood of neonates may be correlated with the severity of encephalopathy and brain injury [57]. The other three trials confirmed these results, while Nagdyman et al. found no differences in the level of NSE in 29 asphyxiated neonates compared with 20 controls [60,61,62,63].

5.4. Butyrylcarnitine

Fatty acid b-oxidation takes place in the mitochondria, involving at least 31 enzymes or the transporters involved. Short-chain acyl-CoA dehydrogenase (SCAD) is the enzyme that catalyzes the first phase of mitochondrial fatty acid β-oxidation. Increased butyrylcarnitine is the result of the dysfunction of this enzyme [93].
A retrospective observational cohort study by Lopez-Suarez et al. analyzed the acetyl-carnitine profile of 67 infants with early HIE in the perinatal period (days 1–7 of life) [62].
The acetyl-carnitine profile is generally measured in neonatal screening for the early detection of inherited metabolic disease by means of electrospray tandem ionization with mass spectrometry coupled to a high-performance liquid chromatography system [94,95].
Lopez-Suarez et al. found a significant positive correlation between butyrylcarnitine and NSE [62]. Therefore, butyrylcarnitine and NSE seems to be the best prognostic biomarkers of neuronal insult in HIE.

5.5. Acidic Calcium-Binding Protein

The acidic calcium-binding protein (S-100B) is a member of a family of calcium binding proteins named S-100 proteins [96]. This protein is localized to some extent in non-neuronal cell types including melanocytes, Langerhans cells, dendritic cells in lymphoid organs, chondrocytes, Leydig cells, adrenal medulla satellite cells, and skeletal muscle satellite cells [97]. S-100 proteins work as calcium sensor proteins that modulate biological activity via calcium binding and perform several cellular mechanisms within the cell populations that contain it such as necrosis and apoptosis [98,99,100]. Additionally, S100B is found in extracellular biological fluids, due to its active secretion from cells [98].
Focusing on the central nervous system, S-100B is expressed and released by astrocytes. This secretion by glia is an early response to metabolic injury (i.e., oxygen, serum, and glucose deprivation) and could be released in biological fluids at an early stage with a renal metabolism [87,101]. The S100B half-life is about one hour. Cord blood and urine seems to be a perfect source to titrate the S100B concentration using LIAISON or ELISA techniques within 2–6 h [68,102].
A number of studies evaluated the ability of S-100B testing to predict brain injury. In this context, Beharier’s study summarizes the available data regarding the sensitivity and specificity values of S100B testing in urine and serum (monitoring time points up to 24 h), showing a sensitivity of 50% to 73% and a specificity of 74% to 90% in serum [97].
This protein plays a trophic role during the development of the nervous system with paracrine/autocrine/endocrine trophic role at low, physiological concentrations (nanomolar). These trophic effects include neuronal survival, muscle development, and the regeneration and promotion of neurite extension [97,103]. However, its overexpression can have dangerous effects due to the activation of inducible NO synthase and the subsequent production of NO with astrocyte cell death [104]. Since the S-100B protein, during an active brain injury, is released from a damaged tissue into circulation, its concentration increases at an early stage of hypoxia in both cerebrospinal fluid and cord blood. For this reason, the best sources for biomarkers are the fluids obtained the least invasively and shortly after birth [105]. Chaparro et al. collected white blood cells isolated from venous blood from children with encephalopathy, using polymerase chain reaction analysis that revealed substantial increases in the expression of S-100B by 97% compared with healthy controls [57]. Serum concentrations of S-100B increase in correlation with the severity of HIE, white matter brain lesions, and this has long-term neurological consequences: in severe asphyxia, the S-100B levels were found to be high immediately after birth and continue to rise with time (up to one week); in mild asphyxia, the blood levels exhibit only a slight elevation soon after birth and decline from this point further with time [61,67,106,107,108,109,110]. Newborns with no signs of asphyxia, instead, present baseline levels of the protein at all times [96].
The data revealed that S-100B testing can discriminate immediately after birth between asphyxiated newborns with a severe clinical outcome and newborns suffering from asphyxia with no clinical outcome. The ability of S100B testing to predict prognosis was in association with the severity of the cases [109]. However, S-100B predictive values have been shown to be less accurate in cases where the clinical outcome is severe but not clearly defined [70]. Interestingly, a marked elevation of the protein was found also in term and preterm asphyxiated newborns complicated with IVH [64,66,107].
Previous studies about S100B concentrations that increased in the case of a brain injury suggested that higher S100B levels in pregnancies with FGR reflect fetal chronic hypoxia [65,106,111]. So, the protein can be used as a biomarker of brain damage in growth-restricted newborns [65,69]. The examination of the cord blood S-100B concentration may be helpful in identifying SGA newborns at a higher risk of postnatal neurological sequelae at an early stage in cases where a prenatal Doppler examination is normal, even when the standard clinical and laboratory parameters are silent, and an early stage neurologic follow-up is uneventful [68]. Gazzolo et al. reported that higher concentrations of S-100B were detected in the mother of FGR fetuses who developed IVH after birth [112]. To satisfy the criteria for the reliable use of S-100B as an accurate screening test, combinations of biomarkers might be needed to improve the outcome prediction. For example, combining S-100B with NSE, a biomarker of early neuronal necrotic damage, may increase the early detection of neuronal damage and expose different patterns of brain damage [60,61,63,96].

5.6. F2-Isoprostanes

F2-Isoprostanes (IPs) are made by the peroxidation of lipids in cell membranes as the result of FRs-induced injury [113]. So, IPs are a useful biomarker for lipid peroxidation. They can be important in clinical practice because preterm white matter is vulnerable to a lipid peroxidation-mediated injury [72].
In preterm infants, during the developmental window between 23 and 32 weeks, the brain is at a significant risk of WMI: the white matter can be exposed to OS due to hypoxia-ischemia [114]. Preoligodendrocytes (OLs) dominate the white matter during this phase [115] and appear particularly prone to FRs-mediated injury because of the immaturity of antioxidant defenses, whereas differentiated OLs are more resistant to OS [116].
Quantifying the IPs in human brain tissue, it can be possible to identify in the glia the aldehydes which originated from lipid hydroperoxides [116].
Coviello et al. showed that the cord blood Ips (cb-Ips) and plasma Ips (pl-Ips) (between 24 and 48 h after birth) levels were not significantly different [72]. Univariate regression analysis demonstrated that cb-IPs were not associated with WMI at the term of an equivalent age and with the cognitive and motor outcome at 24 months of the corrected age. Instead, pl-IPs were positively associated with WMI at the term of an equivalent age; especially, a higher pl-IPs concentration and lower GA were associated with a higher WMI score. In this study, the pl-IPs levels plotted curve indicated that 31.8 pg/mL had the best predictive threshold, with a sensitivity of 86% and a lower specificity of 60% to discriminate newborns with and without WMI, while they were not associated with the cognitive and motor outcome at 24 months of a corrected age [72]. Matthews et al. demonstrated increased pl-IPs in preterm infants at risk of severe abnormalities on neuroimaging [71]. These results revealed that the early pl-IPs concentration was higher in infants with WMI and this correlation remained statistically significant after adjusting for potential confounding factors. Thus, the pl-IPs levels might be a valuable early biomarker of WMI [72]. In addition, Coviello et al. demonstrated a relation between higher pl-IPs levels and a decreased functional brain activity measured with amplitude-integrated EEG [73]. These data are in accordance with the literature according to which IPs are significantly raised in infants with WMI [116].

5.7. Nucleated Red Blood Cells

Another marker of brain damage which has been described is the rise in nucleated red blood cells (NRBC) in the peripheral neonatal blood following birth asphyxia [74]. Fotopoulos’s study showed that at a mean postnatal age of 24 h, the absolute numbers of NRBC in the peripheral blood of asphyxiated neonates were significantly higher compared with those of the controls, while during the following days, the absolute NRBC numbers showed a progressive fall in both groups [59]. In addition, in the same period, the NRBCs numbers were significantly higher in the neonates who developed neurologic abnormalities than in those who had a normal neurologic development [59]. Florio et al. confirmed these results [75].

5.8. Non-Protein-Bound Protein

The term non-protein-bound protein (NPBI) indicates a form of iron free from plasma protein binding and with a low molecular mass. The method for measuring the NPBI levels in small samples is based on the preferential chelation of NPBI by a large excess of low affinity ligand nitrilotriacetic acid through high performance liquid chromatography [117]. Free iron is toxic when not bound to proteins because it is potentially available to produce the hydroxyl radical by reacting with H2O2 through the Fenton reaction, the latter being the main cause of oxidative damage [118]. Brain damage is caused by the absorption of plasma NPBI, which crosses the damaged blood–brain barrier. The high concentrations of plasma NBPI serve the oligodendrocytes in the process of differentiation, thus increasing their susceptibility to OS from FRs. Additionally, the hypoxia and ischemia caused by perinatal asphyxia strongly contributes to the release of NPBI [4,119].
Buonocore et al. showed that no children with normal NBPI values subsequently exhibited neurological abnormalities, showing a 100% sensitivity and 100% specificity for a good neurological outcome. Additionally, a high concentration of NPBI in addition to an exposure to high amounts of lipids leads to the formation of IsoPs, which also cause oxidative damage [76].

5.9. Activina A

Activin is a member of the transforming growth factor β superfamily, a trophic factor that regulates the differentiation and proliferation of a wide variety of cells [120]. Activin A is a neuroprotective factor during brain damage and hypoxic-ischemic damage. Mechanical irritation and chemical brain damage evoke a strong upregulation of activin A [121,122]. Activin A and its receptors are widely distributed throughout the brain. High plasma concentrations of Activin A have been found in both IVH premature infants and term infants with moderate to severe asphyxia, in which cases the activin A analysis reached a sensitivity of 100% and a specificity of 93% as a single marker [75,77,123].

5.10. Erythropoietin

EPO is a glycoprotein hormone produced mainly by the kidneys in response to cellular hypoxia [124,125]. EPO and its receptor are also expressed in astrocytes, neurons, and endothelial cells of the brain [126].
The pilot study by Bhandari et al. demonstrated that high concentrations of EPO were present in the umbilical cord blood of 116 infants under 34 weeks of age with IVH, diagnosed by cUS [78]. The advantage of measuring the EPO concentration in the blood is that it can be measured at birth and the results are available the same day.

5.11. Chemokine Ligand 18

Chemokine ligand 18 (CCL18) is encoded on chromosome 17q11.2 and belongs to the CC chemokine family. It plays a key role in the lymphocyte homing and primary immune response and the CCL18 receptor is detectable in the choroid plexus, periventricular capillary endothelium, ependymal cells, and germline matrix [127].
Therefore, inflammatory conditions can lead to the increased plasma levels of CCL18 [128]. Premature infants who developed CP and patients with traumatic injuries have been shown to have lower levels of CCL18 in umbilical cord blood in the first case and elevated levels of CCL18 in brain tissue biopsies in the second [79,129].
Kallankari et al. measured in 116 premature infants less than 107 umbilical cord blood immunoproteins. Infants who developed IVH shortly after birth had lower concentrations of umbilical cord chemokine CCL18 than very preterm babies without IVH [79]. CCL18 seems to predict the risk of grade II-IV IVH, having ruled out its association with chorioamnionitis or funisitis. High levels of CCL18 block the action of the agonist ligands on CCR3 and thus inhibit the degranulation of leukocyte and, consequently, the inflammatory activity acts as a protective factors against IVH and brain injury [79].

5.12. 24S-Hydroxycholesterol

24S-hydroxycholesterol (24S-HC) is a brain-derived cholesterol metabolite, produced by neuron-specific cytochrome P450 enzyme, CYP46A1, exclusively in the brain and it is capable of crossing the blood–brain barrier into circulation and is excreted in bile [130]. CYP46 converts cholesterol into 24S-HC via hydroxylation and upregulates the cholesterol efflux through the activation of the nuclear transcription factor X. It has been demonstrated that CYP46 is responsible for cholesterol efflux in the brain [131].
Recent works, in animal models, suggested that 24S-HC might be a promising novel lipid biomarker for the extent of HI brain injury [132,133]. A few trials have demonstrated the effects of neonatal encephalopathy on brain cholesterol synthesis and the regulation of the cholesterol metabolism, suggesting not only that its metabolites (specifically those released from the brain into the serum) may potentially act as biomarkers to aid in the identification or severity stratification of hypoxic–ischemic brain injury, but also that the cholesterol pathways may be therapeutic in neonatal encephalopathy [132,133].
The hypothesis of using 24S-HC as an HI brain injury marker is made for several reasons: the most important one is that the HI-induced upregulation of CYP46A1 (in mouse) mediates the increased formation of brain 24S-HC, leading to its elevation in the blood at 6 h and 24 h after HI, suggesting that one mechanism resulting in the decreased cholesterol levels may be cholesterol efflux [132,134].
The activation of CYP46A1 leading to an increase in 24SHC could be due to the effects of increased glutamate and OS, as a result of excitotoxicity, because it has been reported that they can enhance the promoter activity of CYP46A1 [135,136,137].
Fuxin et al. demonstrated that serum 24S-HC could be an acute marker of an HI brain injury if measured within 24 h after the insult (cell death responses peak), but it is of limited value if it is measured beyond this time window [138]. In clinical practice, when the pregnant patients show evidence of acute peripartum or intrapartum hypoxia or the interruption of the placental blood flow, the serum 24S-HC can be measured within 24 h to evaluate the brain injury and provide early prognostic information [138]. The 24S-HC levels can be used in conjunction with other criteria for selecting babies eligible for therapeutic hypothermia within 6 h from their birth [138]. According to these studies in animal models, 24S-HC can be considered an early predictive serum marker for both a diagnostic and prognostic application for infants with HIE.

5.13. Neurotrophins

The nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) are crucial for the development of the peripheral and central nervous system [139]. A growing body of evidence suggests that neurotrophic factors can protect neurons against neuronal death [140,141]. Kirschner et al. examined whether the systemic administration of members of the neurotrophin family, NGF, BDNF, neurotrophin 3, and neurotrophin 5, and the basic fibroblast growth factor (bFGF) could protect against brain damage in neonatal rats [142]. It was demonstrated that neurotrophins and bFGF can attenuate hypoxia-induced neuronal damage by reducing OS.
In an experimental model of hypoxic-ischemic brain damage, inflammatory cytokines such as TNF-α, ICAM-1, and IL-1β contribute to neuronal apoptosis, whereas neurotrophins NGF and BDNF antagonize it [143].
Recently, urinary NGF has been suggested as an early prognostic indicator of a high long-term risk of motor and cognitive impairment in SGA and preterm neonates [144].

6. Placenta and Perinatal Brain Damage

The placenta plays a key role in the plasticity of fetal development. Fetal neurodevelopment is the mirror of the chemical, biochemical, and placental histopathological composition; therefore, placental examination can be a useful tool to identify fetal/neonatal brain damage early and assess its extent [2]. The inflammation and alteration of placental perfusion are associated with higher levels of OS biomarkers in umbilical cord blood. This may indicate an increased fetal susceptibility to oxidative damage [145].
Normal placental functions can be altered by the abundant production of ROS and reactive nitrogen species. Cord blood and the placenta represent important sources of OS biomarkers that can consent to identify early high-risk newborn [2]. Perrone et al. tested the hypothesis that a placental injury is associated with increased levels of OS biomarkers in umbilical cord blood. They analyzed the three different OS markers of isoprostanes, non-protein-bound iron, and advanced oxidative protein products and they found that these markers were detectable in the umbilical cord blood in patients born to mothers with chorioamnionitis and reduced perfusion [145]. The disadvantage of fetal biomarkers is that they do not allow for follow-up studies because more samples are needed in order to monitor the changes throughout time.
Yanni et al. demonstrated that placental inflammation and postnatal systemic inflammation together were associated with a higher risk for white matter damage according to the two-hit hypothesis [146]. The first hit is placental inflammation and the second hit is the elevated concentration of the cytokines (CRP, TNF-α, IL-8, and ICAM-1) in the top quartile for gestational and postnatal age. The synergetic action of the two factors was associated with ventriculomegaly, hypoechoic lesion, and microcephaly [146].
The inflammation of the placenta was also associated with low corticotropin-releasing hormone (CRH) mRNA concentrations. The high activity of placental CRH (identical in structure, immunoreactivity, and bioactivity to hypothalamic CRH) determines much of the increase in free cortisol of the pregnant woman [147,148]. Cortisol is extremely important for brain development in an anatomical and functional sense and very low levels are associated with poor neurodevelopment; although, very high levels negatively affect brain development [149].
Leviton et al. explained that the placenta CRH concentration appears to convey information about the risk of brain damage in extremely preterm newborns. They showed that infants whose placenta had a low concentration of CRH mRNA were at an increased risk of cUS abnormalities such as ventriculomegaly [150].
Stressful events during pregnancy may adversely affect brain development and may increase the risk of neurodevelopmental disorders later in life. It has been assumed that early changes in the kynurenine pathway of tryptophan degradation, which contains quinolinic acid, a neuroactive metabolite, may represent the molecular link between prenatal stress and delayed pathological consequences [151].
All together, these data suggest that endogenous prenatal phenomena influence the risk of brain damage in preterm infants. Further studies on the postnatal evaluation of human placental cellular and molecular mediators could be useful for identifying unfavorable intrauterine conditions, the early stratification of a population of newborns at risk for brain damage, and developing successful intervention/prevention strategies.

7. Conclusions

Not only preterm but also the term brain is particularly vulnerable to the OS-related damage. The presence of an association between biomarkers of oxidative stress measured in the first hours of life and brain damage (successfully evaluated through neuroimaging) emphasizes the possibility of an early identification of newborns at a greater risk of brain damage.
Reliable early specific serum markers, associated with brain damage in newborns, are currently lacking in clinical practice. S-100B seems to be the most studied biomarker. However, the literature is inconsistent due to the complexity of brain damage etiology, the timing of the insult, and blood sampling, limiting the validation for its use in clinical practice. An ideal biomarker, to be useful, should inform about the condition at the time of the measurement for an early treatment and predicts disease progression in order to provide patients with more information on the future clinical outcomes. The ideal biomarker, furthermore, should be easily isolated from the blood, with a low cost, and identify in infants with a brain injury early in the first few hours of their life. In addition, a good biomarker may help to identify the timing of the injury; this is essential because an HI injury often begins in utero, and if too much time has elapsed since the initial brain injury, the neonate will not benefit from treatment. Cut off values are needed to move the biomarkers from the bench to the bedside and the assay must be not only of a high sensitivity but suitable for a very rapid processing and return of the results for the clinician to act on. Finally, the correlation between placental inflammation and brain damage should be evaluated in further studies. It might be useful to identify a panel of placental biomarkers capable of identifying patients at risk of brain damage earlier than neonatal serum biomarkers. For the best prognosis, more studies should focus also on the association of these biomarkers with the type and severity of perinatal brain damage considering the low number of studies that evaluate this relation.

Author Contributions

Conceptualization, S.P., S.M.R.E. and G.B. (Giuseppe Buonocore); methodology, F.G., C.C., G.B. (Giovanni Boscarino) and G.C.; validation, S.P., G.B. (Giuseppe Buonocore) and S.M.R.E.; formal analysis, F.G. and C.C.; investigation, G.B. (Giovanni Boscarino) and G.C.; resources, L.M.G., A.D.P. and S.M.; data curation, S.P.; writing—original draft preparation, F.G., C.C., G.B. (Giovanni Boscarino) and G.C.; writing—review and editing, C.P., L.M.G., A.D.P. and S.M.; visualization, S.P. and S.M.R.E.; supervision, S.P., G.B. (Giuseppe Buonocore) and S.M.R.E.; project administration, S.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Project funded under the National Recovery and Resilience Plan (NRRP), Mission 4 “Education and Research” Component 2 “From research to enterprise” Investment 1.3—funded by the European Union—NextGenerationEU.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

OSoxidative stress
FRsfree radicals
cUScerebral ultrasound
MRImagnetic resonance imaging
IVHintraventricular hemorrhage
PVHIperiventricular hemorrhagic infarction
PVLperiventricular leukomalacia
CPcerebral palsy
HIEhypoxic ischemic encephalopathy
HIhypoxic ischemia
cCMVcongenital cytomegalovirus
DPP4dipeptidylpeptidase 4
NOnitric oxide
ROSreactive oxygen species
NSEneuron-specific enolase
SCADshort-chain acyl-CoA dehydrogenase
S-100Bacidic calcium-binding protein
SGAsmall for gestational age
AGAadequate for gestational age
IPsF2-Isoprostanes
WMIwhite matter injury
OLspreoligodendrocytes
NRBCnucleated red blood cells
NPBInon-protein-bound protein
EPOerythropoietin
CCL18chemokine ligand 18
24S-HC24S-hydroxycholesterol
NGFnerve growth factor
BDNFBrain-derived neurotrophic factor
bFGFbasic fibroblast growth factor
CRHcorticotropin-releasing hormone

References

  1. Tataranno, M.L.; Perrone, S.; Buonocore, G. Plasma Biomarkers of Oxidative Stress in Neonatal Brain Injury. Clin. Perinatol. 2015, 42, 529–539. [Google Scholar] [CrossRef] [PubMed]
  2. Perrone, S.; Santacroce, A.; Picardi, A.; Buonocore, G. Fetal Programming and Early Identification of Newborns at High Risk of Free Radical-Mediated Diseases. WJCP 2016, 5, 172. [Google Scholar] [CrossRef] [PubMed]
  3. Czerska, M.; Mikołajewska, K.; Zieliński, M.; Gromadzińska, J.; Wąsowicz, W. Today’s Oxidative Stress Markers. Med. Pr. 2015, 66, 393–405. [Google Scholar] [CrossRef]
  4. Perrone, S.; Tataranno, L.M.; Stazzoni, G.; Ramenghi, L.; Buonocore, G. Brain Susceptibility to Oxidative Stress in the Perinatal Period. J. Matern.-Fetal Neonatal Med. 2015, 28, 2291–2295. [Google Scholar] [CrossRef]
  5. Lembo, C.; Buonocore, G.; Perrone, S. Oxidative Stress in Preterm Newborns. Antioxidants 2021, 10, 1672. [Google Scholar] [CrossRef] [PubMed]
  6. Bersani, I.; Pluchinotta, F.; Dotta, A.; Savarese, I.; Campi, F.; Auriti, C.; Chuklantseva, N.; Piersigilli, F.; Gazzolo, F.; Varrica, A.; et al. Early Predictors of Perinatal Brain Damage: The Role of Neurobiomarkers. Clin. Chem. Lab. Med. (CCLM) 2020, 58, 471–486. [Google Scholar] [CrossRef] [Green Version]
  7. Volpe, J.J.; Volpe, J.J. (Eds.) Volpe’s Neurology of the Newborn, 6th ed.; Elsevier: Philadelphia, PA, USA, 2018; ISBN 978-0-323-42876-7. [Google Scholar]
  8. Volpe, J.J. Brain Injury in Premature Infants: A Complex Amalgam of Destructive and Developmental Disturbances. Lancet Neurol. 2009, 8, 110–124. [Google Scholar] [CrossRef] [Green Version]
  9. Girard, N.J.; Dory-Lautrec, P.; Koob, M.; Dediu, A.M. MRI Assessment of Neonatal Brain Maturation. Imaging Med. 2012, 4, 613–632. [Google Scholar] [CrossRef]
  10. Kostović, I.; Radoš, M.; Kostović-Srzentić, M.; Krsnik, Ž. Fundamentals of the Development of Connectivity in the Human Fetal Brain in Late Gestation: From 24 Weeks Gestational Age to Term. J. Neuropathol. Exp. Neurol. 2021, 80, 393–414. [Google Scholar] [CrossRef]
  11. Andersen, A.; Fisker, A.B.; Rodrigues, A.; Martins, C.; Ravn, H.; Lund, N.; Biering-Sørensen, S.; Benn, C.S.; Aaby, P. National Immunization Campaigns with Oral Polio Vaccine Reduce All-Cause Mortality: A Natural Experiment within Seven Randomized Trials. Front. Public Health 2018, 6, 13. [Google Scholar] [CrossRef]
  12. Boscarino, G.; Conti, M.G.; Gasparini, C.; Onestà, E.; Faccioli, F.; Dito, L.; Regoli, D.; Spalice, A.; Parisi, P.; Terrin, G. Neonatal Hyperglycemia Related to Parenteral Nutrition Affects Long-Term Neurodevelopment in Preterm Newborn: A Prospective Cohort Study. Nutrients 2021, 13, 1930. [Google Scholar] [CrossRef]
  13. Puia-Dumitrescu, M.; Comstock, B.A.; Li, S.; Heagerty, P.J.; Perez, K.M.; Law, J.B.; Wood, T.R.; Gogcu, S.; Mayock, D.E.; Juul, S.E.; et al. Assessment of 2-Year Neurodevelopmental Outcomes in Extremely Preterm Infants Receiving Opioids and Benzodiazepines. JAMA Netw. Open 2021, 4, e2115998. [Google Scholar] [CrossRef] [PubMed]
  14. Boscarino, G.; Di Chiara, M.; Cellitti, R.; De Nardo, M.C.; Conti, M.G.; Parisi, P.; Spalice, A.; Di Mario, C.; Ronchi, B.; Russo, A.; et al. Effects of Early Energy Intake on Neonatal Cerebral Growth of Preterm Newborn: An Observational Study. Sci. Rep. 2021, 11, 18457. [Google Scholar] [CrossRef] [PubMed]
  15. Khurana, S.; Kane, A.E.; Brown, S.E.; Tarver, T.; Dusing, S.C. Effect of Neonatal Therapy on the Motor, Cognitive, and Behavioral Development of Infants Born Preterm: A Systematic Review. Dev. Med. Child Neurol. 2020, 62, 684–692. [Google Scholar] [CrossRef] [PubMed]
  16. Terrin, G.; Boscarino, G.; Gasparini, C.; Di Chiara, M.; Faccioli, F.; Onestà, E.; Parisi, P.; Spalice, A.; De Nardo, M.C.; Dito, L.; et al. Energy-Enhanced Parenteral Nutrition and Neurodevelopment of Preterm Newborns: A Cohort Study. Nutrition 2021, 89, 111219. [Google Scholar] [CrossRef] [PubMed]
  17. Terrin, G.; De Nardo, M.C.; Boscarino, G.; Di Chiara, M.; Cellitti, R.; Ciccarelli, S.; Gasparini, C.; Parisi, P.; Urna, M.; Ronchi, B.; et al. Early Protein Intake Influences Neonatal Brain Measurements in Preterms: An Observational Study. Front. Neurol. 2020, 11, 885. [Google Scholar] [CrossRef]
  18. Whitelaw, A. Intraventricular Haemorrhage and Posthaemorrhagic Hydrocephalus: Pathogenesis, Prevention and Future Interventions. Semin. Neonatol. 2001, 6, 135–146. [Google Scholar] [CrossRef]
  19. Deng, W.; Pleasure, J.; Pleasure, D. Progress in Periventricular Leukomalacia. Arch. Neurol. 2008, 65, 1291–1295. [Google Scholar] [CrossRef] [Green Version]
  20. Bracci, R.; Perrone, S.; Buonocore, G. The Timing of Neonatal Brain Damage. Neonatology 2006, 90, 145–155. [Google Scholar] [CrossRef]
  21. Perrone, S.; Lembo, C.; Gironi, F.; Petrolini, C.; Catalucci, T.; Corbo, G.; Buonocore, G.; Gitto, E.; Esposito, S.M.R. Erythropoietin as a Neuroprotective Drug for Newborn Infants: Ten Years after the First Use. Antioxidants 2022, 11, 652. [Google Scholar] [CrossRef]
  22. Kolnik, S.; Wood, T.R. Role of Vitamin E in Neonatal Neuroprotection: A Comprehensive Narrative Review. Life 2022, 12, 1083. [Google Scholar] [CrossRef] [PubMed]
  23. Abiramalatha, T.; Ramaswamy, V.V.; Ponnala, A.K.; Kallem, V.R.; Murkunde, Y.V.; Punnoose, A.M.; Vivekanandhan, A.; Pullattayil, A.K.; Amboiram, P. Emerging Neuroprotective Interventions in Periventricular Leukomalacia—A Systematic Review of Preclinical Studies. Expert Opin. Investig. Drugs 2022, 31, 305–330. [Google Scholar] [CrossRef] [PubMed]
  24. A Report: The Definition and Classification of Cerebral Palsy April 2006. Dev. Med. Child Neurol. 2007, 49, 8–14. [CrossRef]
  25. The Australian Cerebral Palsy Register. Australian Cerebral Palsy Register Bulletin: Birth Years 1995–2014; The Australian Cerebral Palsy Register: North Adelaide, Australia, 2020. [Google Scholar]
  26. MacLennan, A.H.; Thompson, S.C.; Gecz, J. Cerebral Palsy: Causes, Pathways, and the Role of Genetic Variants. Am. J. Obstet. Gynecol. 2015, 213, 779–788. [Google Scholar] [CrossRef] [Green Version]
  27. Novak, I.; Morgan, C.; Adde, L.; Blackman, J.; Boyd, R.N.; Brunstrom-Hernandez, J.; Cioni, G.; Damiano, D.; Darrah, J.; Eliasson, A.-C.; et al. Early, Accurate Diagnosis and Early Intervention in Cerebral Palsy: Advances in Diagnosis and Treatment. JAMA Pediatr. 2017, 171, 897. [Google Scholar] [CrossRef]
  28. Doyle, L.W.; Crowther, C.A.; Middleton, P.; Marret, S.; Rouse, D. Magnesium Sulphate for Women at Risk of Preterm Birth for Neuroprotection of the Fetus. Cochrane Database Syst. Rev. 2009, 21, CD004661. [Google Scholar] [CrossRef]
  29. Schmidt, B.; Roberts, R.S.; Davis, P.; Doyle, L.W.; Barrington, K.J.; Ohlsson, A.; Solimano, A.; Tin, W. Long-Term Effects of Caffeine Therapy for Apnea of Prematurity. N. Engl. J. Med. 2007, 357, 1893–1902. [Google Scholar] [CrossRef] [Green Version]
  30. Spittle, A.J.; Morgan, C.; Olsen, J.E.; Novak, I.; Cheong, J.L.Y. Early Diagnosis and Treatment of Cerebral Palsy in Children with a History of Preterm Birth. Clin. Perinatol. 2018, 45, 409–420. [Google Scholar] [CrossRef]
  31. Hamilton, B.E.; Hoyert, D.L.; Martin, J.A.; Strobino, D.M.; Guyer, B. Annual Summary of Vital Statistics: 2010–2011. Pediatrics 2013, 131, 548–558. [Google Scholar] [CrossRef] [Green Version]
  32. Yıldız, E.P.; Ekici, B.; Tatlı, B. Neonatal Hypoxic Ischemic Encephalopathy: An Update on Disease Pathogenesis and Treatment. Expert Rev. Neurother. 2017, 17, 449–459. [Google Scholar] [CrossRef]
  33. Shankaran, S.; Laptook, A.R.; Ehrenkranz, R.A.; Tyson, J.E.; McDonald, S.A.; Donovan, E.F.; Fanaroff, A.A.; Poole, W.K.; Wright, L.L.; Higgins, R.D.; et al. Whole-Body Hypothermia for Neonates with Hypoxic–Ischemic Encephalopathy. N. Engl. J. Med. 2005, 353, 1574–1584. [Google Scholar] [CrossRef] [PubMed]
  34. Thoresen, M.; Tooley, J.; Liu, X.; Jary, S.; Fleming, P.; Luyt, K.; Jain, A.; Cairns, P.; Harding, D.; Sabir, H. Time Is Brain: Starting Therapeutic Hypothermia within Three Hours after Birth Improves Motor Outcome in Asphyxiated Newborns. Neonatology 2013, 104, 228–233. [Google Scholar] [CrossRef] [PubMed]
  35. Azzopardi, D.V.; Strohm, B.; Edwards, A.D.; Dyet, L.; Halliday, H.L.; Juszczak, E.; Kapellou, O.; Levene, M.; Marlow, N.; Porter, E.; et al. Moderate Hypothermia to Treat Perinatal Asphyxial Encephalopathy. N. Engl. J. Med. 2009, 361, 1349–1358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ferriero, D.M. Neonatal Brain Injury. N. Engl. J. Med. 2004, 351, 1985–1995. [Google Scholar] [CrossRef] [PubMed]
  37. Albrecht, M.; Zitta, K.; Groenendaal, F.; van Bel, F.; Peeters-Scholte, C. Neuroprotective Strategies Following Perinatal Hypoxia-Ischemia: Taking Aim at NOS. Free. Radic. Biol. Med. 2019, 142, 123–131. [Google Scholar] [CrossRef]
  38. Bennet, L.; Roelfsema, V.; Pathipati, P.; Quaedackers, J.S.; Gunn, A.J. Relationship between Evolving Epileptiform Activity and Delayed Loss of Mitochondrial Activity after Asphyxia Measured by Near-Infrared Spectroscopy in Preterm Fetal Sheep: Post-Hypoxic Mitochondrial Failure in Preterm Fetal Sheep. J. Physiol. 2006, 572, 141–154. [Google Scholar] [CrossRef]
  39. Cross, J.L.; Meloni, B.P.; Bakker, A.J.; Lee, S.; Knuckey, N.W. Modes of Neuronal Calcium Entry and Homeostasis Following Cerebral Ischemia. Stroke Res. Treat. 2010, 2010, 1–9. [Google Scholar] [CrossRef] [Green Version]
  40. Cerio, F.; Lara-Celador, I.; Alvarez, A.; Hilario, E. Neuroprotective Therapies after Perinatal Hypoxic-Ischemic Brain Injury. Brain Sci. 2013, 3, 191–214. [Google Scholar] [CrossRef] [PubMed]
  41. Nelson, K.B.; Lynch, J.K. Stroke in Newborn Infants. Lancet Neurol. 2004, 3, 150–158. [Google Scholar] [CrossRef] [Green Version]
  42. Laugesaar, R.; Kolk, A.; Tomberg, T.; Metsvaht, T.; Lintrop, M.; Varendi, H.; Talvik, T. Acutely and Retrospectively Diagnosed Perinatal Stroke: A Population-Based Study. Stroke 2007, 38, 2234–2240. [Google Scholar] [CrossRef]
  43. Lee, J. Maternal and Infant Characteristics Associated With Perinatal Arterial Stroke in the Infant. JAMA 2005, 293, 723. [Google Scholar] [CrossRef] [PubMed]
  44. Kirton, A.; deVeber, G. Life After Perinatal Stroke. Stroke 2013, 44, 3265–3271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Bernson-Leung, M.E.; Rivkin, M.J. Stroke in Neonates and Children. Pediatr. Rev. 2016, 37, 463–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Arai, K.; Jin, G.; Navaratna, D.; Lo, E.H. Brain Angiogenesis in Developmental and Pathological Processes: Neurovascular Injury and Angiogenic Recovery after Stroke: Neurovascular Responses in Stroke. FEBS J. 2009, 276, 4644–4652. [Google Scholar] [CrossRef] [Green Version]
  47. Kirton, A. Advancing Non-Invasive Neuromodulation Clinical Trials in Children: Lessons from Perinatal Stroke. Eur. J. Paediatr. Neurol. 2017, 21, 75–103. [Google Scholar] [CrossRef] [PubMed]
  48. Wanigasinghe, J.; Reid, S.M.; Mackay, M.T.; Reddihough, D.S.; Harvey, A.S.; Freeman, J.L. Epilepsy in Hemiplegic Cerebral Palsy Due to Perinatal Arterial Ischaemic Stroke: Epilepsy in CP Due to Stroke. Dev. Med. Child Neurol. 2010, 52, 1021–1027. [Google Scholar] [CrossRef]
  49. Boppana, S.B.; Rivera, L.B.; Fowler, K.B.; Mach, M.; Britt, W.J. Intrauterine Transmission of Cytomegalovirus to Infants of Women with Preconceptional Immunity. N. Engl. J. Med. 2001, 344, 1366–1371. [Google Scholar] [CrossRef] [PubMed]
  50. Pass, R.F.; Fowler, K.B.; Boppana, S.B.; Britt, W.J.; Stagno, S. Congenital Cytomegalovirus Infection Following First Trimester Maternal Infection: Symptoms at Birth and Outcome. J. Clin. Virol. 2006, 35, 216–220. [Google Scholar] [CrossRef]
  51. Natale, F.; De Curtis, M.; Bizzarri, B.; Orlando, M.P.; Ralli, M.; Liuzzi, G.; Caravale, B.; Franco, F.; Gaeta, A.; Giancotti, A.; et al. Isolated Auditory Neuropathy at Birth in Congenital Cytomegalovirus Infection. Ital. J. Pediatr. 2020, 46, 3. [Google Scholar] [CrossRef] [Green Version]
  52. Thigpen, J. Congenital Cytomegalovirus—History, Current Practice, and Future Opportunities. Neonatal Netw. 2020, 39, 293–298. [Google Scholar] [CrossRef]
  53. Johnson, J.; Anderson, B. Cytomegalovirus: Should We Screen Pregnant Women for Primary Infection? Am. J. Perinatol. 2013, 30, 121–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chiopris, G.; Veronese, P.; Cusenza, F.; Procaccianti, M.; Perrone, S.; Daccò, V.; Colombo, C.; Esposito, S. Congenital Cytomegalovirus Infection: Update on Diagnosis and Treatment. Microorganisms 2020, 8, 1516. [Google Scholar] [CrossRef] [PubMed]
  55. Tomicic, M.T.; Bey, E.; Wutzler, P.; Thust, R.; Kaina, B. Comparative Analysis of DNA Breakage, Chromosomal Aberrations and Apoptosis Induced by the Anti-Herpes Purine Nucleoside Analogues Aciclovir, Ganciclovir and Penciclovir. Mutat. Res. /Fundam. Mol. Mech. Mutagen. 2002, 505, 1–11. [Google Scholar] [CrossRef]
  56. Yakovleva, A.A.; Zolotov, N.N.; Sokolov, O.Y.; Kost, N.V.; Kolyasnikova, K.N.; Micheeva, I.G. Dipeptidylpeptidase 4 (DPP4, CD26) Activity in the Blood Serum of Term and Preterm Neonates with Cerebral Ischemia. Neuropeptides 2015, 52, 113–117. [Google Scholar] [CrossRef] [PubMed]
  57. Chaparro-Huerta, V.; Flores-Soto, M.E.; Merin Sigala, M.E.; Barrera de León, J.C.; Lemus-Varela, M.d.L.; Torres-Mendoza, B.M.d.G.; Beas-Zárate, C. Proinflammatory Cytokines, Enolase and S-100 as Early Biochemical Indicators of Hypoxic-Ischemic Encephalopathy Following Perinatal Asphyxia in Newborns. Pediatr. Neonatol. 2017, 58, 70–76. [Google Scholar] [CrossRef] [PubMed]
  58. Chiesa, C.; Pellegrini, G.; Panero, A.; De Luca, T.; Assumma, M.; Signore, F.; Pacifico, L. Umbilical Cord Interleukin-6 Levels Are Elevated in Term Neonates with Perinatal Asphyxia: Serum IL-6 after Perinatal Asphyxia. Eur. J. Clin. Investig. 2003, 33, 352–358. [Google Scholar] [CrossRef] [PubMed]
  59. Fotopoulos, S.; Pavlou, K.; Skouteli, H.; Papassotiriou, I.; Lipsou, N.; Xanthou, M. Early Markers of Brain Damage in Premature Low-Birth-Weight Neonates Who Suffered from Perinatal Asphyxia and/or Infection. Biol. Neonate 2001, 79, 213–218. [Google Scholar] [CrossRef]
  60. Nagdyman, N.; Kömen, W.; Ko, H.K.; Müller, C.; Obladen, M. Early Biochemical Indicators of Hypoxic-Ischemic Encephalopathy after Birth Asphyxia. Pediatr. Res. 2001, 49, 502–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Giuseppe, D.; Sergio, C.; Pasqua, B.; Giovanni, L.V.; Salvatore, C.; Frigiola, A.; Petra, H.; Maurizio, A. Perinatal Asphyxia in Preterm Neonates Leads to Serum Changes in Protein S-100 and Neuron Specific Enolase. Curr. Neurovasc. Res. 2009, 6, 110–116. [Google Scholar] [CrossRef]
  62. López-Suárez, O.; Concheiro-Guisán, A.; Sánchez-Pintos, P.; Cocho, J.A.; Fernández Lorenzo, J.R.; Couce, M.L. Acylcarnitine Profile in Neonatal Hypoxic-Ischemic Encephalopathy: The Value of Butyrylcarnitine as a Prognostic Marker. Medicine 2019, 98, e15221. [Google Scholar] [CrossRef]
  63. 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. [Google Scholar] [CrossRef]
  64. Gazzolo, D.; Di Iorio, R.; Marinoni, E.; Masetti, P.; Serra, G.; Giovannini, L.; Michetti, F. S100B Protein Is Increased in Asphyxiated Term Infants Developing Intraventricular Hemorrhage. Crit. Care Med. 2002, 30, 1356–1360. [Google Scholar] [CrossRef]
  65. Gazzolo, D.; Marinoni, E.; Di Iorio, R.; Lituania, M.; Bruschettini, P.L.; Michetti, F. Circulating S100β Protein Is Increased in Intrauterine Growth-Retarded Fetuses. Pediatr. Res. 2002, 51, 215–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Gazzolo, D.; Vinesi, P.; Bartocci, M.; Geloso, M.C.; Bonacci, W.; Serra, G.; Haglid, K.G.; Michetti, F. Elevated S100 Blood Level as an Early Indicator of Intraventricular Hemorrhage in Preterm Infants. Correlation with Cerebral Doppler Velocimetry. J. Neurol. Sci. 1999, 170, 32–35. [Google Scholar] [CrossRef] [PubMed]
  67. Murabayashi, M.; Minato, M.; Okuhata, Y.; Makimoto, M.; Hosono, S.; Masaoka, N.; Okada, T.; Yamamoto, T.; Mugishima, H.; Takahashi, S.; et al. Kinetics of Serum S100B in Newborns with Intracranial Lesions. Pediatr. Int. 2008, 50, 17–22. [Google Scholar] [CrossRef]
  68. Strzalko, B.; Karowicz-Bilinska, A.; Wyka, K.; Krajewski, P.; Kesiak, M.; Kociszewska-Najman, B. Serum S100B Protein Concentrations in SGA/FGR Newborns. Ginekol. Pol. 2022, 93, 158–162. [Google Scholar] [CrossRef] [PubMed]
  69. Velipaşaoğlu, M.; Yurdakök, M.; Özyüncü, Ö.; Portakal, O.; Deren, Ö. Neural Injury Markers to Predict Neonatal Complications in Intrauterine Growth Restriction. J. Obstet. Gynaecol. 2015, 35, 555–560. [Google Scholar] [CrossRef] [PubMed]
  70. Thorngren-Jerneck, K.; Alling, C.; Herbst, A.; Amer-Wahlin, I.; Marsal, K. S100 Protein in Serum as a Prognostic Marker for Cerebral Injury in Term Newborn Infants with Hypoxic Ischemic Encephalopathy. Pediatr. Res. 2004, 55, 406–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Matthews, M.A.; Aschner, J.L.; Stark, A.R.; Moore, P.E.; Slaughter, J.C.; Steele, S.; Beller, A.; Milne, G.L.; Settles, O.; Chorna, O.; et al. Increasing F2-Isoprostanes in the First Month after Birth Predicts Poor Respiratory and Neurodevelopmental Outcomes in Very Preterm Infants. J. Perinatol. 2016, 36, 779–783. [Google Scholar] [CrossRef] [Green Version]
  72. Coviello, C.; Perrone, S.; Buonocore, G.; Negro, S.; Longini, M.; Dani, C.; de Vries, L.S.; Groenendaal, F.; Vijlbrief, D.C.; Benders, M.J.N.L.; et al. Isoprostanes as Biomarker for White Matter Injury in Extremely Preterm Infants. Front. Pediatr. 2020, 8, 618622. [Google Scholar] [CrossRef]
  73. Coviello, C.; Perrone, S.; Buonocore, G.; Negro, S.; Longini, M.; Groenendaal, F.; Vijlbrief, D.C.; Dani, C.; Benders, M.J.N.L.; Tataranno, M.L. Oxidative Stress Biomarkers and Early Brain Activity in Extremely Preterm Infants: A Prospective Cohort Study. Children 2022, 9, 1376. [Google Scholar] [CrossRef] [PubMed]
  74. Green, D.W.; Hendon, B.; Mimouni, F.B. Nucleated Erythrocytes and Intraventricular Hemorrhage in Preterm Neonates. Pediatrics 1995, 96, 475–478. [Google Scholar] [CrossRef] [PubMed]
  75. Florio, P.; Perrone, S.; Luisi, S.; Longini, M.; Tanganelli, D.; Petraglia, F.; Buonocore, G. Activin A Plasma Levels at Birth: An Index of Fetal Hypoxia in Preterm Newborn. Pediatr. Res. 2003, 54, 696–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Buonocore, G.; Perrone, S.; Longini, M.; Paffetti, P.; Vezzosi, P.; Gatti, M.G.; Bracci, R. Non Protein Bound Iron as Early Predictive Marker of Neonatal Brain Damage. Brain 2003, 126, 1224–1230. [Google Scholar] [CrossRef] [Green Version]
  77. Florio, P.; Perrone, S.; Luisi, S.; Vezzosi, P.; Longini, M.; Marzocchi, B.; Petraglia, F.; Buonocore, G. Increased Plasma Concentrations of Activin A Predict Intraventricular Hemorrhage in Preterm Newborns. Clin. Chem. 2006, 52, 1516–1521. [Google Scholar] [CrossRef] [Green Version]
  78. Bhandari, V.; Buhimschi, C.S.; Han, C.S.; Lee, S.Y.; Pettker, C.M.; Campbell, K.H.; Dulay, A.T.; Oliver, E.A.; Werner, E.F.; Buhimschi, I.A. Cord Blood Erythropoietin and Interleukin-6 for Prediction of Intraventricular Hemorrhage in the Preterm Neonate. J. Matern.-Fetal Neonatal Med. 2011, 24, 673–679. [Google Scholar] [CrossRef] [Green Version]
  79. Kallankari, H.; Kaukola, T.; Ojaniemi, M.; Herva, R.; Perhomaa, M.; Vuolteenaho, R.; Kingsmore, S.F.; Hallman, M. Chemokine CCL18 Predicts Intraventricular Hemorrhage in Very Preterm Infants. Ann. Med. 2010, 42, 416–425. [Google Scholar] [CrossRef]
  80. Lambeir, A.-M.; Durinx, C.; Scharpé, S.; De Meester, I. Dipeptidyl-Peptidase IV from Bench to Bedside: An Update on Structural Properties, Functions, and Clinical Aspects of the Enzyme DPP IV. Crit. Rev. Clin. Lab. Sci. 2003, 40, 209–294. [Google Scholar] [CrossRef]
  81. Ansorge, S.; Reinhold, D. Immune Peptides Related to Dipeptidyl Aminopeptidase IV/CD26. In Handbook of Biologically Active Peptides; Elsevier: Amsterdam, The Netherlands, 2006; pp. 567–572. ISBN 978-0-12-369442-3. [Google Scholar]
  82. García-Horsman, J.A.; Männistö, P.T.; Venäläinen, J.I. On the Role of Prolyl Oligopeptidase in Health and Disease. Neuropeptides 2007, 41, 1–24. [Google Scholar] [CrossRef]
  83. Röhnert, P.; Schmidt, W.; Emmerlich, P.; Goihl, A.; Wrenger, S.; Bank, U.; Nordhoff, K.; Täger, M.; Ansorge, S.; Reinhold, D.; et al. Dipeptidyl Peptidase IV, Aminopeptidase N and DPIV/APN-like Proteases in Cerebral Ischemia. J. Neuroinflamm. 2012, 9, 44. [Google Scholar] [CrossRef]
  84. Lokshina, L.A. Plasma membrane proteinases from lymphoid cells and their biological functions. Bioorg. Khim. 1998, 24, 323–331. [Google Scholar] [PubMed]
  85. Malva, J.O.; Xapelli, S.; Baptista, S.; Valero, J.; Agasse, F.; Ferreira, R.; Silva, A.P. Multifaces of Neuropeptide Y in the Brain—Neuroprotection, Neurogenesis and Neuroinflammation. Neuropeptides 2012, 46, 299–308. [Google Scholar] [CrossRef] [PubMed]
  86. Iwai, T.; Sawabe, T.; Tanimitsu, K.; Suzuki, M.; Sasaki-Hamada, S.; Oka, J.-I. Glucagon-like Peptide-1 Protects Synaptic and Learning Functions from Neuroinflammation in Rodents: GLP-1 Prevents Inflammation-Induced Dysfunction. J. Neurosci. Res. 2014, 92, 446–454. [Google Scholar] [CrossRef] [PubMed]
  87. Douglas-Escobar, M.; Weiss, M.D. Biomarkers of Hypoxic-Ischemic Encephalopathy in Newborns. Front. Neur. 2012, 3, 144. [Google Scholar] [CrossRef] [Green Version]
  88. Algra, S.O.; Groeneveld, K.M.; Schadenberg, A.W.; Haas, F.; Evens, F.C.; Meerding, J.; Koenderman, L.; Jansen, N.J.; Prakken, B.J. Cerebral Ischemia Initiates an Immediate Innate Immune Response in Neonates during Cardiac Surgery. J Neuroinflamm. 2013, 10, 796. [Google Scholar] [CrossRef] [Green Version]
  89. Cowell, R.M.; Xu, H.; Galasso, J.M.; Silverstein, F.S. Hypoxic-Ischemic Injury Induces Macrophage Inflammatory Protein-1α Expression in Immature Rat Brain. Stroke 2002, 33, 795–801. [Google Scholar] [CrossRef] [Green Version]
  90. Kaur, C.; Rathnasamy, G.; Ling, E.-A. Roles of Activated Microglia in Hypoxia Induced Neuroinflammation in the Developing Brain and the Retina. J. Neuroimmune Pharmacol. 2013, 8, 66–78. [Google Scholar] [CrossRef]
  91. Leviton, A. Preterm Birth and Cerebral Palsy: Is Tumor Necrosis Factor the Missing Link? Dev. Med. Child Neurol. 2008, 35, 553–558. [Google Scholar] [CrossRef]
  92. Isgrò, M.A.; Bottoni, P.; Scatena, R. Neuron-Specific Enolase as a Biomarker: Biochemical and Clinical Aspects. In Advances in Cancer Biomarkers; Scatena, R., Ed.; Advances in Experimental Medicine and Biology; Springer: Dordrecht, The Netherlands, 2015; Volume 867, pp. 125–143. ISBN 978-94-017-7214-3. [Google Scholar]
  93. Wolfe, L.; Jethva, R.; Oglesbee, D.; Vockley, J. Short-Chain Acyl-CoA Dehydrogenase Deficiency. In GeneReviews®; Adam, M.P., Everman, D.B., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Bean, L.J., Gripp, K.W., Amemiya, A., Eds.; University of Washington Seattle: Seattle, WA, USA, 1993. [Google Scholar]
  94. Fernandez-Lainez, C.; Aguilar-Lemus, J.; Vela-Amieva, M.; Ibarra-Gonzalez, I. Tandem Mass Spectrometry Newborn Screening for Inborn Errors of Intermediary Metabolism: Abnormal Profile Interpretation. CMC 2012, 19, 4511–4522. [Google Scholar] [CrossRef]
  95. Millington, D.S.; Kodo, N.; Norwood, D.L.; Roe, C.R. Tandem Mass Spectrometry: A New Method for Acylcarnitine Profiling with Potential for Neonatal Screening for Inborn Errors of Metabolism. J Inher. Metab. Dise. 1990, 13, 321–324. [Google Scholar] [CrossRef]
  96. Beharier, O.; Kahn, J.; Shusterman, E.; Sheiner, E. S100B—A Potential Biomarker for Early Detection of Neonatal Brain Damage Following Asphyxia. J. Matern.-Fetal Neonatal Med. 2012, 25, 1523–1528. [Google Scholar] [CrossRef] [PubMed]
  97. Michetti, F.; Corvino, V.; Geloso, M.C.; Lattanzi, W.; Bernardini, C.; Serpero, L.; Gazzolo, D. The S100B Protein in Biological Fluids: More than a Lifelong Biomarker of Brain Distress. J. Neurochem. 2012, 120, 644–659. [Google Scholar] [CrossRef] [PubMed]
  98. Ikura, M. Calcium Binding and Conformational Response in EF-Hand Proteins. Trends Biochem. Sci. 1996, 21, 14–17. [Google Scholar] [CrossRef] [PubMed]
  99. Donato, R.; Sorci, G.; Riuzzi, F.; Arcuri, C.; Bianchi, R.; Brozzi, F.; Tubaro, C.; Giambanco, I. S100B’s Double Life: Intracellular Regulator and Extracellular Signal. Biochim. Biophys. Acta 2009, 1793, 1008–1022. [Google Scholar] [CrossRef] [Green Version]
  100. Van Eldik, L.J.; Wainwright, M.S. The Janus Face of Glial-Derived S100B: Beneficial and Detrimental Functions in the Brain. Restor. Neurol. Neurosci. 2003, 21, 97–108. [Google Scholar] [PubMed]
  101. Gerlach, R.; Demel, G.; König, H.-G.; Gross, U.; Prehn, J.H.M.; Raabe, A.; Seifert, V.; Kögel, D. Active Secretion of S100B from Astrocytes during Metabolic Stress. Neuroscience 2006, 141, 1697–1701. [Google Scholar] [CrossRef]
  102. Gazzolo, D.; Abella, R.; Marinoni, E.; di Iorio, R.; Li Volti, G.; Galvano, F.; Frigiola, A.; Temporini, F.; Moresco, L.; Colivicchi, M.; et al. New Markers of Neonatal Neurology. J. Matern Fetal Neonatal Med. 2009, 22 (Suppl. 3), 57–61. [Google Scholar] [CrossRef]
  103. Gazzolo, D.; Michetti, F. Perinatal S100B Protein Assessment in Human Unconventional Biological Fluids: A Minireview and New Perspectives. Cardiovasc. Psychiatry Neurol. 2010, 2010, 703563. [Google Scholar] [CrossRef] [Green Version]
  104. Lam, A.G.; Koppal, T.; Akama, K.T.; Guo, L.; Craft, J.M.; Samy, B.; Schavocky, J.P.; Watterson, D.M.; Van Eldik, L.J. Mechanism of Glial Activation by S100B: Involvement of the Transcription Factor NFkappaB. Neurobiol. Aging 2001, 22, 765–772. [Google Scholar] [CrossRef]
  105. Florio, P. Biochemical Markers of Perinatal Brain Damage. Front. Biosci. 2010, S2, 47–72. [Google Scholar] [CrossRef]
  106. Gazzolo, D.; Abella, R.; Marinoni, E.; Di Iorio, R.; Li Volti, G.; Galvano, F.; Pongiglione, G.; Frigiola, A.; Bertino, E.; Florio, P. Circulating Biochemical Markers of Brain Damage in Infants Complicated by Ischemia Reperfusion Injury. Cardiovasc. Hematol. Agents Med. Chem. 2009, 7, 108–126. [Google Scholar] [CrossRef] [PubMed]
  107. 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, 1836–1838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Gazzolo, D.; Marinoni, E.; Di Iorio, R.; Bruschettini, M.; Kornacka, M.; Lituania, M.; Majewska, U.; Serra, G.; Michetti, F. Measurement of Urinary S100B Protein Concentrations for the Early Identification of Brain Damage in Asphyxiated Full-Term Infants. Arch. Pediatr. Adolesc. Med. 2003, 157, 1163–1168. [Google Scholar] [CrossRef] [PubMed]
  109. Gazzolo, D.; Frigiola, A.; Bashir, M.; Iskander, I.; Mufeed, H.; Aboulgar, H.; Venturini, P.; Marras, M.; Serra, G.; Frulio, R.; et al. Diagnostic Accuracy of S100B Urinary Testing at Birth in Full-Term Asphyxiated Newborns to Predict Neonatal Death. PLoS ONE 2009, 4, e4298. [Google Scholar] [CrossRef]
  110. Gazzolo, D.; Marinoni, E.; Di Iorio, R.; Bruschettini, M.; Kornacka, M.; Lituania, M.; Majewska, U.; Serra, G.; Michetti, F. Urinary S100B Protein Measurements: A Tool for the Early Identification of Hypoxic-Ischemic Encephalopathy in Asphyxiated Full-Term Infants. Crit. Care Med. 2004, 32, 131–136. [Google Scholar] [CrossRef]
  111. Tskitishvili, E.; Komoto, Y.; Temma-Asano, K.; Hayashi, S.; Kinugasa, Y.; Tsubouchi, H.; Song, M.; Kanagawa, T.; Shimoya, K.; Murata, Y. S100B Protein Expression in the Amnion and Amniotic Fluid in Pregnancies Complicated by Pre-Eclampsia. Mol. Hum. Reprod. 2006, 12, 755–761. [Google Scholar] [CrossRef] [Green Version]
  112. Gazzolo, D.; Marinoni, E.; Di Iorio, R.; Lituania, M.; Marras, M.; Bruschettini, M.; Bruschettini, P.; Frulio, R.; Michetti, F.; Petraglia, F.; et al. High Maternal Blood S100B Concentrations in Pregnancies Complicated by Intrauterine Growth Restriction and Intraventricular Hemorrhage. Clin. Chem. 2006, 52, 819–826. [Google Scholar] [CrossRef] [Green Version]
  113. Milne, G.L.; Musiek, E.S.; Morrow, J.D. F2-Isoprostanes as Markers of Oxidative Stress in Vivo: An Overview. Biomarkers 2005, 10, 10–23. [Google Scholar] [CrossRef]
  114. Ferriero, D.M.; Miller, S.P. Imaging Selective Vulnerability in the Developing Nervous System: Imaging Selective Vulnerability. J. Anat. 2010, 217, 429–435. [Google Scholar] [CrossRef]
  115. Back, S.A.; Luo, N.L.; Borenstein, N.S.; Levine, J.M.; Volpe, J.J.; Kinney, H.C. Late Oligodendrocyte Progenitors Coincide with the Developmental Window of Vulnerability for Human Perinatal White Matter Injury. J. Neurosci. 2001, 21, 1302–1312. [Google Scholar] [CrossRef]
  116. Back, S.A.; Luo, N.L.; Mallinson, R.A.; O’Malley, J.P.; Wallen, L.D.; Frei, B.; Morrow, J.D.; Petito, C.K.; Roberts, C.T.; Murdoch, G.H.; et al. Selective Vulnerability of Preterm White Matter to Oxidative Damage Defined by F2-Isoprostanes. Ann. Neurol. 2005, 58, 108–120. [Google Scholar] [CrossRef] [PubMed]
  117. Paffetti, P.; Perrone, S.; Longini, M.; Ferrari, A.; Tanganelli, D.; Marzocchi, B.; Buonocore, G. Non-Protein-Bound Iron Detection in Small Samples of Biological Fluids and Tissues. BTER 2006, 112, 221–232. [Google Scholar] [CrossRef] [PubMed]
  118. Papanikolaou, G.; Pantopoulos, K. Iron Metabolism and Toxicity. Toxicol. Appl. Pharmacol. 2005, 202, 199–211. [Google Scholar] [CrossRef]
  119. Buonocore, G.; Perrone, S.; Bracci, R. Free Radicals and Brain Damage in the Newborn. Biol. Neonate 2001, 79, 180–186. [Google Scholar] [CrossRef]
  120. Florio, P.; Gazzolo, D.; Luisi, S.; Petraglia, F. Activin A In Brain Injury. In Advances in Clinical Chemistry; Elsevier: Amsterdam, The Netherlands, 2007; Volume 43, pp. 117–130. ISBN 978-0-12-373703-8. [Google Scholar]
  121. He, J.-T.; Mang, J.; Mei, C.-L.; Yang, L.; Wang, J.-Q.; Xing, Y.; Yang, H.; Xu, Z.-X. Neuroprotective Effects of Exogenous Activin A on Oxygen-Glucose Deprivation in PC12 Cells. Molecules 2011, 17, 315–327. [Google Scholar] [CrossRef] [PubMed]
  122. Mukerji, S.S.; Katsman, E.A.; Wilber, C.; Haner, N.A.; Selman, W.R.; Hall, A.K. Activin Is a Neuronal Survival Factor That Is Rapidly Increased after Transient Cerebral Ischemia and Hypoxia in Mice. J. Cereb. Blood Flow Metab. 2007, 27, 1161–1172. [Google Scholar] [CrossRef] [Green Version]
  123. Florio, P.; Luisi, S.; Bruschettini, M.; Grutzfeld, D.; Dobrzanska, A.; Bruschettini, P.; Petraglia, F.; Gazzolo, D. Cerebrospinal Fluid Activin A Measurement in Asphyxiated Full-Term Newborns Predicts Hypoxic Ischemic Encephalopathy. Clin. Chem. 2004, 50, 2386–2389. [Google Scholar] [CrossRef]
  124. Davis, L.E.; Widness, J.A.; Brace, R.A. Renal and Placental Secretion of Erythropoietin during Anemia or Hypoxia in the Ovine Fetus. Am. J. Obstet. Gynecol. 2003, 189, 1764–1770. [Google Scholar] [CrossRef]
  125. Teramo, K.A.; Widness, J.A. Increased Fetal Plasma and Amniotic Fluid Erythropoietin Concentrations: Markers of Intrauterine Hypoxia. Neonatology 2009, 95, 105–116. [Google Scholar] [CrossRef]
  126. Marti, H.H. Erythropoietin and the Hypoxic Brain. J. Exp. Biol. 2004, 207, 3233–3242. [Google Scholar] [CrossRef]
  127. Zlotnik, A.; Yoshie, O.; Nomiyama, H. The Chemokine and Chemokine Receptor Superfamilies and Their Molecular Evolution. Genome Biol. 2006, 7, 243. [Google Scholar] [CrossRef] [PubMed]
  128. Schutyser, E. Involvement of CC Chemokine Ligand 18 (CCL18) in Normal and Pathological Processes. J. Leukoc. Biol. 2005, 78, 14–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Chang, C.-Y.; Lee, Y.-H.; Leu, S.-J.; Wang, C.-Y.; Wei, C.-P.; Hung, K.-S.; Pai, M.-H.; Tsai, M.-D.; Wu, C.-H. CC-Chemokine Ligand 18/Pulmonary Activation-Regulated Chemokine Expression in the CNS with Special Reference to Traumatic Brain Injuries and Neoplastic Disorders. Neuroscience 2010, 165, 1233–1243. [Google Scholar] [CrossRef] [PubMed]
  130. Björkhem, I.; Lütjohann, D.; Diczfalusy, U.; Ståhle, L.; Ahlborg, G.; Wahren, J. Cholesterol Homeostasis in Human Brain: Turnover of 24S-Hydroxycholesterol and Evidence for a Cerebral Origin of Most of This Oxysterol in the Circulation. J. Lipid Res. 1998, 39, 1594–1600. [Google Scholar] [CrossRef]
  131. Cartagena, C.M.; Burns, M.P.; Rebeck, G.W. 24S-Hydroxycholesterol Effects on Lipid Metabolism Genes Are Modeled in Traumatic Brain Injury. Brain Res. 2010, 1319, 1–12. [Google Scholar] [CrossRef] [Green Version]
  132. Lu, F.; Zhu, J.; Guo, S.; Wong, B.J.; Chehab, F.F.; Ferriero, D.M.; Jiang, X. Upregulation of Cholesterol 24-Hydroxylase Following Hypoxia-Ischemia in Neonatal Mouse Brain. Pediatr. Res. 2018, 83, 1218–1227. [Google Scholar] [CrossRef] [Green Version]
  133. Yu, Z.; Li, S.; Lv, S.H.; Piao, H.; Zhang, Y.H.; Zhang, Y.M.; Ma, H.; Zhang, J.; Sun, C.K.; Li, A.P. Hypoxia-Ischemia Brain Damage Disrupts Brain Cholesterol Homeostasis in Neonatal Rats. Neuropediatrics 2009, 40, 179–185. [Google Scholar] [CrossRef]
  134. Dave, A.M.; Peeples, E.S. Cholesterol Metabolism and Brain Injury in Neonatal Encephalopathy. Pediatr. Res. 2021, 90, 37–44. [Google Scholar] [CrossRef]
  135. Mast, N.; Anderson, K.W.; Johnson, K.M.; Phan, T.T.N.; Guengerich, F.P.; Pikuleva, I.A. In Vitro Cytochrome P450 46A1 (CYP46A1) Activation by Neuroactive Compounds. J. Biol. Chem. 2017, 292, 12934–12946. [Google Scholar] [CrossRef] [Green Version]
  136. Ohyama, Y.; Meaney, S.; Heverin, M.; Ekström, L.; Brafman, A.; Shafir, M.; Andersson, U.; Olin, M.; Eggertsen, G.; Diczfalusy, U.; et al. Studies on the Transcriptional Regulation of Cholesterol 24-Hydroxylase (CYP46A1): Marked Insensitivity toward Different Regulatory Axes. J. Biol. Chem. 2006, 281, 3810–3820. [Google Scholar] [CrossRef]
  137. Sodero, A.O.; Weissmann, C.; Ledesma, M.D.; Dotti, C.G. Cellular Stress from Excitatory Neurotransmission Contributes to Cholesterol Loss in Hippocampal Neurons Aging in Vitro. Neurobiol. Aging 2011, 32, 1043–1053. [Google Scholar] [CrossRef]
  138. Lu, F.; Fan, S.; Romo, A.R.; Xu, D.; Ferriero, D.M.; Jiang, X. Serum 24S-Hydroxycholesterol Predicts Long-Term Brain Structural and Functional Outcomes after Hypoxia-Ischemia in Neonatal Mice. J. Cereb. Blood Flow Metab. 2021, 41, 312–323. [Google Scholar] [CrossRef] [PubMed]
  139. Skaper, S.D. Neurotrophic Factors: An Overview. In Neurotrophic Factors; Skaper, S.D., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2018; Volume 1727, pp. 1–17. ISBN 978-1-4939-7570-9. [Google Scholar]
  140. Sun, Z.; Hu, W.; Yin, S.; Lu, X.; Zuo, W.; Ge, S.; Xu, Y. NGF Protects against Oxygen and Glucose Deprivation-Induced Oxidative Stress and Apoptosis by up-Regulation of HO-1 through MEK/ERK Pathway. Neurosci. Lett. 2017, 641, 8–14. [Google Scholar] [CrossRef]
  141. Kowiański, P.; Lietzau, G.; Czuba, E.; Waśkow, M.; Steliga, A.; Moryś, J. BDNF: A Key Factor with Multipotent Impact on Brain Signaling and Synaptic Plasticity. Cell Mol. Neurobiol. 2018, 38, 579–593. [Google Scholar] [CrossRef]
  142. Kirschner, P.B.; Jenkins, B.G.; Schulz, J.B.; Finkelstein, S.P.; Matthews, R.T.; Rosen, B.R.; Flint Beal, M. NGF, BDNF and NT-5, but Not NT-3 Protect against MPP+ Toxicity and Oxidative Stress in Neonatal Animals. Brain Res. 1996, 713, 178–185. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, Y.; Cao, M.; Liu, A.; Di, W.; Zhao, F.; Tian, Y.; Jia, J. Changes of Inflammatory Cytokines and Neurotrophins Emphasized Their Roles in Hypoxic–Ischemic Brain Damage. Int. J. Neurosci. 2013, 123, 191–195. [Google Scholar] [CrossRef] [PubMed]
  144. Aisa, M.C.; Barbati, A.; Cappuccini, B.; De Rosa, F.; Gerli, S.; Clerici, G.; Kaptilnyy, V.A.; Ishenko, A.I.; Di Renzo, G.C. Urinary Nerve Growth Factor in Full-Term, Preterm and Intra Uterine Growth Restriction Neonates: Association with Brain Growth at 30–40 Days of Postnatal Period and with Neuro-Development Outcome at Two Years. A Pilot Study. Neurosci. Lett. 2021, 741, 135459. [Google Scholar] [CrossRef] [PubMed]
  145. Perrone, S.; Tataranno, M.L.; Negro, S.; Longini, M.; Toti, M.S.; Alagna, M.G.; Proietti, F.; Bazzini, F.; Toti, P.; Buonocore, G. Placental Histological Examination and the Relationship with Oxidative Stress in Preterm Infants. Placenta 2016, 46, 72–78. [Google Scholar] [CrossRef]
  146. Yanni, D.; Korzeniewski, S.J.; Allred, E.N.; Fichorova, R.N.; O’Shea, T.M.; Kuban, K.; Dammann, O.; Leviton, A. for the ELGAN Study Investigators; Both Antenatal and Postnatal Inflammation Contribute Information about the Risk of Brain Damage in Extremely Preterm Newborns. Pediatr. Res. 2017, 82, 691–696. [Google Scholar] [CrossRef] [Green Version]
  147. Hagberg, H.; Mallard, C.; Ferriero, D.M.; Vannucci, S.J.; Levison, S.W.; Vexler, Z.S.; Gressens, P. The Role of Inflammation in Perinatal Brain Injury. Nat. Rev. Neurol. 2015, 11, 192–208. [Google Scholar] [CrossRef]
  148. Dammann, O.; Leviton, A. Intermittent or Sustained Systemic Inflammation and the Preterm Brain. Pediatr. Res. 2014, 75, 376–380. [Google Scholar] [CrossRef] [PubMed]
  149. Vohr, B.R.; Wright, L.L.; Poole, W.K.; McDonald, S.A.; for the NICHD Neonatal Research Network Follow-up Study. Neurodevelopmental Outcomes of Extremely Low Birth Weight Infants <32 Weeks’ Gestation Between 1993 and 1998. Pediatrics 2005, 116, 635–643. [Google Scholar] [CrossRef] [PubMed]
  150. Leviton, A.; Allred, E.N.; Kuban, K.C.K.; O’Shea, T.M.; Paneth, N.; Majzoub, J.; ELGAN Study Investigators. Brain Disorders Associated with Corticotropin-Releasing Hormone Expression in the Placenta among Children Born before the 28th Week of Gestation. Acta Paediatr. 2016, 105, e7–e11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Notarangelo, F.M.; Schwarcz, R. Restraint Stress during Pregnancy Rapidly Raises Kynurenic Acid Levels in Mouse Placenta and Fetal Brain. Dev. Neurosci. 2016, 38, 458–468. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematically representation of IVH classification by Volpe. Figure legend: PVHI (periventricular hemorrhagic infarction).
Figure 1. Schematically representation of IVH classification by Volpe. Figure legend: PVHI (periventricular hemorrhagic infarction).
Antioxidants 12 00309 g001
Figure 2. Newborn brain damage and serum biomarkers. Figure legend: IVH (intraventricular hemorrhage); PLV (periventricular leukomalacia); CP (cerebral palsy); HIE (hypoxic ischemic encephalopathy); DPP4 (dipeptidylpeptidase 4); NSE (neuron-specific enolase); S-100B (acidic calcium-binding protein); IPs (F2-isoprostanes); NRBC (nucleated red blood cells); NPBI (non-protein-bound protein); EPO (erythropoietin); CCL18 (chemokine ligand 18); 24S-HC (24S-hydroxycholesterol).
Figure 2. Newborn brain damage and serum biomarkers. Figure legend: IVH (intraventricular hemorrhage); PLV (periventricular leukomalacia); CP (cerebral palsy); HIE (hypoxic ischemic encephalopathy); DPP4 (dipeptidylpeptidase 4); NSE (neuron-specific enolase); S-100B (acidic calcium-binding protein); IPs (F2-isoprostanes); NRBC (nucleated red blood cells); NPBI (non-protein-bound protein); EPO (erythropoietin); CCL18 (chemokine ligand 18); 24S-HC (24S-hydroxycholesterol).
Antioxidants 12 00309 g002
Table 1. Summary of the studies that evaluated the relation between serum biomarkers and brain damage in newborns.
Table 1. Summary of the studies that evaluated the relation between serum biomarkers and brain damage in newborns.
BiomarkerReferencePopulationSample Source (i.e., Cord or Peripheral Blood)Brain Damage Patterns
DPP4Yakovleva et al. [56]39 full-term and 40 pretermCord and peripheral blood (3–5 days of life and before discharge)↑ cerebral ischemia
CytokinesChaparro-Huerta et al. [57]62 full-term newborns (32 control vs. 30 Asphyxiated)Plasma samples in the first 24 h of lifeTNF-α, IL-1β and IL-6
↑ in HIE
Chiesa et al. [58]Full-term: 50 case and 113 controlsCord bloodIL-6 ↑ in HIE and worse neurodevelopmental outcome
Fotopoulos et al. [59]57 LBW newbornsPeripheral bloodTNF-α, IL-1β and IL-6
↑ Asphyxiated/infected
NSEChaparro-Huerta et al. [57]62 full-term newborns (32 control vs. 30 Asphyxiated)Plasma samples in the first 24 h of life↑ in HIE
Nagdyman et al. [60]29 asphyxiate and 20 control infantscord blood and plasma samples at 2, 6, 12, and 24 h after birthNo difference
Giuseppe et al. [61]30 neonates with perinatal asphyxia and 10 controlCapillary blood↑ in Perinatal Asphyxia
López-Suárez et al. [62]67 full-term newborns with HIE, 827 full-term controlSerum samples collected in the first
6 h of life and on the 2nd and 3rd day of life
↑ in HIE
Celtik et al. [63]91 full-term newbornsPeripheral blood↑ in HIE
ButyrylcarnitineLópez-Suárez et al. [62]67 full-term newborns with HIE, 827 full-term controlSerum samples collected in the first
6 h of life and on the 2nd and 3rd day of life
↑ in HIE
S-100BNagdyman et al. [60]29 asphyxiated and 20 control infantsCord blood and plasma samples at 2, 6, 12, and 24 h after birth↑ in HIE
Giuseppe et al. [61]30 neonates with perinatal asphyxia and 10 controlCapillary blood↑ in Perinatal Asphyxia
Gazzolo et al. [64]29 full-term newborns with IVH, 20 asphyxiated infants without IVH, and 80 normal newborns.Plasma sample at 12 h of life↑ in IVH
Gazzolo et al. [65]10 IUGR with normal and 10 with abnormal umbilical artery doppler findings. 40 uncomplicated pregnanciesCord blood↑ in IUGR, positive correlation with middle cerebral artery pulsatility index and with umbilical artery pulsatility index to middle cerebral artery pulsatility index ratio.
Gazzolo et al. [66]11 IVH and 14 controlsPlasma samples in the first 72 h of life↑ in IVH, correlated with the grade of hemorrhage. Positive correlation with the middle cerebral artery pulsatility index and S-100B.
Murabayashi et al. [67]22 normal and 40 newborns with brain diseasesSerum samples on day 1, 2 and 6↑ in HIE and Asphyxia
Chaparro-Huerta et al. [57]62 full-term newborns (32 control vs. 30 Asphyxiated)Plasma samples in the first 24 h of life↑ in HIE
Strzalko et al. [68]88 SGA vs. 80 AGACord blood↑ in SGA
Velipaşaoğlu et al. [69]32 IUGR vs. 29 controlsCord blood↑ in IUGR
Thorngren-Jerneck et al. [70]62 full-term infants with birth asphyxiaSerum samples in the first 2–3 days of life↑ in relation with the grade of HIE and ↑ in CP
IPsMatthews et al. [71]136 preterm ≤ 28 wksPlasma samples on days 14 and 28 of life↑ in worse developmental outcomes at 12 months
Coviello et al. [72]44 preterm < 28 wksCord and peripheral blood (24–48 h of life)↑ in WMI
Coviello et al. [73]39 preterm < 28 wksCord and peripheral blood (24–48 h of life)↑ in poor EEG activity
NRBCGreen et al. [74]149 pretermPeripheral blood first 6 days of life↑ IVH
Fotopoulos et al. [59]57 LBW newbornsPeripheral blood↑ Asphyxiated/infected
Florio et al. [75]50 pretermCord blood↑ Hypoxic
NPBIBuonocore et al. [76]384 newborns (225 >36 weeks, 90 from 32 to 36 weeks and 69 <32 wks).Cord blood↑ in worse neurodevelopmental outcome
Activina AFlorio et al. [75]50 pretermCord blood↑ Hypoxic
Florio et al. [77]53 pretermArterial blood in the 1st h of life↑ IVH
EPOBhandari et al. [78]116 pretermCord blood↑ IVH
CCL18Kallankari et al. [79]163 preterm <32 wks.Cord blood↓ IVH
24S-HC No study in human babies, only animals.
NGFAisa et al.43 preterm >32 wks and full-term neonates.No plasma or serum,
only urine.
↓ in worse developmental outcomes at 24 months
Table legend: DPP4 (dipeptidylpeptidase 4); HIE (hypoxic ischemic encephalopathy); NSE (neuron-specific enolase); S-100B (acidic calcium-binding protein); IUGR (intrauterine growth restriction); IVH (intraventricular hemorrhage); SGA (small for gestational age); AGA (adequate for gestational age); CP (cerebral palsy); IPs (F2-isoprostanes); WMI (white matter injury); NRBC (nucleated red blood cells); NPBI (non-protein-bound protein); EPO (erythropoietin); CCL18 (chemokine ligand 18); 24S-HC (24S-hydroxycholesterol), NGF (nerve growth factor). ↑: increase; ↓: decrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Perrone, S.; Grassi, F.; Caporilli, C.; Boscarino, G.; Carbone, G.; Petrolini, C.; Gambini, L.M.; Di Peri, A.; Moretti, S.; Buonocore, G.; et al. Brain Damage in Preterm and Full-Term Neonates: Serum Biomarkers for the Early Diagnosis and Intervention. Antioxidants 2023, 12, 309. https://doi.org/10.3390/antiox12020309

AMA Style

Perrone S, Grassi F, Caporilli C, Boscarino G, Carbone G, Petrolini C, Gambini LM, Di Peri A, Moretti S, Buonocore G, et al. Brain Damage in Preterm and Full-Term Neonates: Serum Biomarkers for the Early Diagnosis and Intervention. Antioxidants. 2023; 12(2):309. https://doi.org/10.3390/antiox12020309

Chicago/Turabian Style

Perrone, Serafina, Federica Grassi, Chiara Caporilli, Giovanni Boscarino, Giulia Carbone, Chiara Petrolini, Lucia Maria Gambini, Antonio Di Peri, Sabrina Moretti, Giuseppe Buonocore, and et al. 2023. "Brain Damage in Preterm and Full-Term Neonates: Serum Biomarkers for the Early Diagnosis and Intervention" Antioxidants 12, no. 2: 309. https://doi.org/10.3390/antiox12020309

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop