Open access peer-reviewed chapter

Preterm Birth and Postnatal Developmental Outcomes

Written By

Jamila Gurbanova, Saadat Huseynova and Afat Hasanova

Submitted: 10 September 2022 Reviewed: 14 September 2022 Published: 02 November 2022

DOI: 10.5772/intechopen.108061

From the Edited Volume

Maternal and Child Health

Edited by Miljana Z. Jovandaric and Sandra Babic

Chapter metrics overview

95 Chapter Downloads

View Full Metrics

Abstract

Premature birth is a pathological condition that requires high-quality medical care due to the infants’ low body mass and gestational age, as well as morphofunctional immaturity. Moreover, such children are at great risk for retardation of mental development; metabolic, cardiovascular, and malignant diseases; and many other health problems at a later age. Early and late complications of preterm birth depend significantly on the gestational age at birth and the intrauterine development conditions of the fetus. Due to the more severe and complicated course of perinatal pathologies, premature babies with fetal growth retardation syndrome constitute a larger risk group. Approximately 50–70% of these children receive long-term treatment in the neonatal intensive care unit after birth. Furthermore, 70% of them face behavioral and memory problems in later life. While the pathologies of the neonatal period in children born prematurely are mainly related to respiratory, gastrointestinal, neurological, and nutritional problems, the complications of premature birth are manifested in children’s early age, preschool, school, adolescence, and other developmental periods.

Keywords

  • preterm birth
  • neurodevelopmental disorders
  • perinatal encephalopathy
  • brain injury
  • perinatal prognostic indicators

1. Introduction

Premature birth is a pathological condition that requires high-quality medical care due to the infants’ low body mass and gestational age, as well as morphofunctional immaturity. Moreover, such children are at great risk for retardation of mental development; metabolic, cardiovascular, and malignant diseases; and many other health problems at a later age [1, 2]. Early and late complications of preterm birth depend significantly on the gestational age at birth and the intrauterine development conditions of the fetus [3, 4]. Due to the more severe and complicated course of perinatal pathologies, premature babies with fetal growth retardation syndrome constitute a larger risk group. Approximately 50–70% of these children receive long-term treatment in the neonatal intensive care unit after birth. Furthermore, 70% of them face behavioral and memory problems in later life [5, 6].

Children with extreme small mass comprise a significant risk group due to developmental and behavioral anomalies. The main problems faced by these children in the neonatal period are related to nutrition and infection [7]. Thus, not only satisfying the energy demand, but also the oral and parenteral intake of the necessary nutrients and microelements leads to the prevention of a number of psychosocial problems at later ages [8, 9]. Additionally, the correct use of antibiotics, the timely implementation of measures to protect against hospital bacterial strains, and the minimization of damage to the integrity of the skin and mucous membranes lead to the reduction of infectious complications [10, 11]. One of the factors leading to the violation of early adaptation in children born prematurely is the immaturity of the painful stimuli response. It is believed that because gestational age and the degree of morphofunctional maturity of the body are low, the cumulative reaction formed in the body against pain stimulus causes not only behavioral disorders but also dysfunctional changes in all organs and systems [12, 13].

Premature birth is the most important medical and social problem of modern times, accounting for 12% of all live births [14]. The rate of prematurity in child mortality varies according to the level of neonatal care in a given region [15]. According to the Institute of Children’s Health and Development, this problem ranks first among causes of child mortality in the African-American zone and second in the Caucasian zone [15, 16]. Despite the improvement of preventive and treatment measures in this field, the fact that the frequency of premature birth is increasing all over the world can be explained by the low level of neonatal care in some regions and the increase in the number of multi-fetal pregnancies as a result of the widespread use of assisted reproductive technologies in developed countries [17, 18]. Statistical studies have shown that even in developed countries, pregnancy ending with premature birth has increased from 9 to 9.4% to 12.7–14% compared to the 1980s [1, 19]. An estimated 75% of the 500,000 preterm births that occur worldwide each year are between 34 and 36 weeks of gestational age, with 25% at a lower gestational age [20, 21, 22]. Studies have shown that 50% of all preterm births occur spontaneously without any specific cause, 40% are medically induced, and 10% occur due to the premature rupture of fetal membranes [23].

Among the risk factors leading to premature birth, in recent years, a history of complicated incomplete birth, multiple pregnancy, and short cervical length have become increasingly relevant. It is believed that women whose previous pregnancies ended prematurely have a higher risk of stillbirth. The recurrent risk depends on the gestational age of the child born during the previous pregnancy and the number of premature births in the history of the woman. Thus, in 50–70% of cases, incomplete birth is repeated at the same gestational age. As well, the risk of repeated preterm birth is 16% in women who have given birth to one child at a gestational age of less than 35 weeks and 41–67% in women who have had multiple pregnancies [24, 25]. Due to the widespread use of assisted reproductive technologies, the number of multiple pregnancies has increased significantly in the last 20 years. Although in vitro fertilization is more successful at achieving and sustaining pregnancy, most twin and multiple pregnancies result in preterm birth [26, 27]. A number of studies have confirmed the association of short cervix size with preterm birth. Some authors have even proposed predicting the time of childbirth based on this indicator [28, 29]. Randomized clinical trials have shown that vaginal progesterone treatment to reduce the risk of premature termination of pregnancy with short cervical length had a positive effect in 45% of cases [25, 30].

Investigations into the possible causes of spontaneous births have found that the intrauterine inflammatory process, mother’s psychosocial stress, fetal psychological stress, uterine overstretching, and uterine bleeding are of significant note [31, 32, 33, 34]. These causes manifest themselves with different frequency depending on the duration of pregnancy. For example, births stimulated by intrauterine bacterial colonization usually occur at a gestational age of less than 34 weeks, while pregnancies that end due to psychosocial problems occur at a gestational age of 34–36 weeks. Preeclampsia, fetal distress, and retardation of fetal development are the main pregnancy pathologies that cause premature birth. According to the data of the International Institute for Child Health and Development, 40% of premature births due to medical intervention are related to women with preeclampsia [35, 36, 37]. Premature rupture of fetal membranes occurs mainly as a result of exacerbation of chronic inflammation, accounting for 21% and 8% of early and late preterm births, respectively [38]. Studies have shown that placental inflammation caused by the impaired protective capacity of the antioxidant protein heme oxygenase-1 plays a fundamental role in the pathogenesis of both preeclampsia and the premature rupture of fetal membranes. Therefore, some scientific studies have been conducted on the effectiveness of drugs with heme oxygenase activity to prolong pregnancy when there is a risk of premature birth [39, 40]. Parturition is a complex biological process regulated by numerous signaling molecules and biologically active substances from the mother, mate, and fetal tissue. A number of preventive measures are implemented to identify and prevent the risk of premature birth. Studies have shown that the placental corticotropin-releasing hormone is the main endocrine mediator of preterm labor [41, 42]. Determination of this factor in pregnancies with a gestation period of less than 33 weeks can be a prognostic indicator of the level of fetal development and premature birth [43, 44, 45]. In recent years, the use of special portable monitors that monitor children’s activity at home has been proposed, but this method is not widely used due to the high economic costs required. The dynamic monitoring of women to prevent any problems that may occur during pregnancy, tocolytic therapy to reduce uterine contractions if necessary, and antibiotic therapy in case of infectious complications are believed to be more effective. Of course, since women whose previous pregnancies ended in premature birth are at greater risk, they should be monitored more actively. The most successful results in the prevention of recurrent premature births have been obtained via the administration of 12 alpha-hydroxyprogesterone injections. Thus, prescribing this drug to women whose previous pregnancies ended in premature birth at 16–20 weeks of gestation significantly reduces the risk of premature birth [46, 47, 48].

Fetal inflammatory response is the main pathogenetic link to preterm birth. The process is due to the influence of microorganisms and is accompanied by chorioamnionitis, funisitis, or intraamnial inflammation [49, 50]. Acute intraventricular hemorrhage, periventricular leukomalacia (PVL), necrotic enterocolitis, bronchopulmonary dysplasia, myocardial dysfunction, and sepsis are more common during the inflammatory process of the fetoplacental system [51, 52, 53, 54]. During the fetal inflammatory response, preterm birth is associated with the effects of cytokines, matrix metalloproteinases, and prostaglandins. In cases of high bacterial colonization, the levels of TNF-α, IL-6, and IL-8 in umbilical cord blood increase significantly [55, 56]. The main clinical manifestations of inflammatory changes that lead to premature birth are uterine prolapse, changes in the cervix, and the premature rupture of the fetal membranes. Unfortunately, during the inflammatory process occurring in the fetoplacental system, the risk of similar inflammation in the mother’s body is very low. Therefore, it is not possible to predict preterm birth based on the inflammatory indicators in the peripheral blood of pregnant women. The cultivation of microbial colonization determined during the examination of the contents taken from the birth canal also does not provide an idea of ​​the level of intraamniotic inflammation [32, 57, 58]. The decrease in the probability of premature birth against the background of antibiotic therapy is also explained by the inhibition of the bacterial infectious process due to the effect of antibacterial drugs [59]. However, there are conflicting and controversial opinions on the use of antibiotic therapy to reduce the probability of premature termination of pregnancy [60, 61, 62]. A group of studies has shown that routine treatment of asymptomatic microbial invasion reduces perinatal morbidity and mortality but has no effect on the incidence of preterm birth [63, 64, 65]. Other studies have shown that bacterial colonization increases the risk of premature birth, although it is not manifested by a specific infectious process. In cervical and vaginal secretions, a membrane protein called fetal fibronectin is found at a frequency 50 times higher in women with the possibility of premature birth than in those with normal pregnancies [66, 67]. The Maternal-Fetal Medicine Union states that the reduction of fetal fibronectin does not occur against the background of antibiotic therapy [68, 69, 70]. As well, despite the weakening of intrauterine colonization against the background of antibacterial therapy, against the background of the use of broad-spectrum antibiotics, no significant improvement in spontaneous births or perinatal and neonatal outcomes has been shown [62, 70]. Therefore, for more accurate diagnosis and data, the contents obtained directly from the fetal membranes rather than from the birth canal should be examined. Elevation of IL-6 and matrix metalloproteinase 8 and 9 levels in amniotic fluid is also considered a more sensitive indicator for amniotic infection [71, 72]. However, because obtaining amniotic fluid is invasive and poses a risk for subsequent infection, this diagnostic method is not widely used. In recent years, it has been considered more appropriate to differentiate inflammatory processes in the amniotic cavity, chorion, and placenta through vaginal cervicometry [73, 74]. Thus, when intraamniotic microbial colonization increases, in 80% of cases, the length of the cervix is ​​shortened, which is called cervical insufficiency [75, 76, 77]. This method is safer and is now widely used in predicting the occurrence of premature birth. Another group of studies has shown that fetal damage during microbial colonization depends on the activity of anti-inflammatory factors in the fetoplacental system. It was determined that as a result of the effect of oxidative stress products, the activation of specific soluble and transmembrane RAGE receptors occurs in macrophages, monocytes, and endothelial cells [78]. This directly leads to the activation of cytokine and growth factor genes [79, 80]. The activation of RAGE receptors during the acceleration of the inflammatory process have been proven to have a protective nature, causing the inhibition of inflammation [81]. Another group of studies has shown that the progress and activity of the inflammatory process depends on the degree of activation of the RAGE and Toll-like receptors [82, 83]. Taketoshi Noguchi and co-authors showed that activation of Toll-like receptors induces preterm labor by leading to a more acute inflammatory process in the fetoplacental system [84]. The activation of RAGE receptors causes the inhibition of the acute process, and the process becomes chronic [85, 86]. A group of studies proved that the polymorphism of Toll-like receptors and the genetic structure of different alleles play an important role in the occurrence of premature birth [87, 88]. Hopefully, large-scale scientific research is being conducted on the genetic basis of the occurrence of premature birth. According to the obtained results thus far, premature pregnancy depends on cytokine, Toll-like receptors, and RAGE polymorphism regulated at the genetic level. Depending on this polymorphism, the inflammatory response of the fetoplacental system is formed. This response, in turn, not only causes premature birth but also significantly affects the course of perinatal pathologies [89]. Uteroplacental ischemia is primarily formed during the inflammatory process in the mother-couple-fetus system [89, 90]. At this time, generalized endothelial dysfunction leads to premature termination of pregnancy and various intrauterine developmental pathologies of the fetus [91]. The pathogenetic mechanisms of the formation of endothelial dysfunction during uteroplacental ischemia have not yet been fully investigated. However, numerous scientific studies have proven that inflammatory processes occurring in the fetoplacental system are often accompanied by thrombophilic conditions [1, 92, 93]. Thrombophilia factors that lead to premature birth are grouped into two categories—congenital and acquired. Congenital thrombophilia is mainly caused by the Leiden mutation and coagulation factor II - prothrombin mutation [94]. Antiphospholipid syndrome is the main acquired factor [95]. The violation of placental microcirculation during thrombophilia can cause thromboembolism, intrauterine death of the fetus, intrauterine growth retardation, premature separation of the pair, acute preeclampsia, and spontaneous abortions [5296, 97].

In recent scientific literature, there have been many reports on the increased incidence of preterm birth in women with mutations of the methylenetetrahydrofolate reductase gene and disorders of folic acid metabolism [98]. It is believed that as a result of mutation, folic acid cannot be activated and transformed into tetrahydrafolate-metafolin. As the latter provides the remethylation of homocysteine, its deficiency leads to disruption of homocysteine transformation and its excessive accumulation in the body [99]. Hyperhomocysteinemia causes generalized damage to the endothelium, atherosclerotic changes, placental microcirculation disorders, premature birth, sudden intrauterine death, as well as congenital developmental anomalies of the neural tube, heart, and a number of organs [100, 101, 102].

One of the main reasons for premature termination of pregnancy is the improper adjustment of the contraction capacity of the uterine muscle depending on the period of gestation. Contraction of the uterus is regulated by the interaction of actin and myosin proteins in myocytes [103]. Myocytes, which create special connections, ensure the synchrony and coordination of contractions of the uterus. The interaction of the actin and myosin complex is realized by the phosphorylation of myosin by the enzyme myosinkinase. The homeostasis of calcium ions plays a key role in myocyte activity. The increase in calcium ions inside the cell is carried out through membrane receptors stimulated by ovarian and placental steroids. Progesterone activates beta adrenergic receptors, and estradiol activates alpha adrenergic, cholinergic, and prostaglandin receptors [104, 105]. The passage of calcium ions into the cell membrane is carried out by alpha adrenergic receptor agonists, and its return is carried out by beta adrenergic receptor agonists. The contraction and relaxation of the uterine muscle depends on the entry of calcium ions from the sarcoplasmic reticulum into the cytoplasm and the amount of agonists and antagonists of multiple receptors involved in this process [106, 107]. Depending on their balance, inadequate contraction of the uterine muscle, loss of synchrony prompts the onset of premature labor. It has been established that uterine contractions during childbirth and labor pains are initiated through the expression of contraction-related protein genes [108, 109]. These genes regulate the synthesis of Connexin-43, the main protein of ion channels and receptors [110]. The role of a number of factors in the regulation of the synchrony of uterine contractions has been indicated—notably, progesterone, nitric oxide, relaxin, prostacyclin, and the corticotropin-releasing hormone (CRH) [110, 111, 112]. These biologically active substances, called hestagens, prevent the release of calcium ions from the sarcoplasmic reticulum by inhibiting cyclic AMF inside the cell. Since numerous experimental studies have shown that these compounds reduce inadequate uterine contractions, they are now widely used in clinical practice to prevent premature birth.

There are many theories about the immunological incompatibility between the mother and the fetus in the etiopathogenesis of premature birth. Increased tolerance of the mother to the fetus depends on the balance of the histological compatibility of complex antigens. While class I HLA-A and HLA-B antigens can be inactivated by the trophoblast, HLA-G antigens expressed during pregnancy protect the fetus from the maternal immune response [102, 113]. Inadequate recognition of fetal antigens by the mother can lead to miscarriage. In animal studies, reduction of galectin-1, a specific immunoregulatory protein, has been shown to stimulate preterm labor [114]. Additionally, in the case of a number of autoimmune diseases (systemic lupus erythematosus, ulcerative colitis, immunopathologies of the thyroid gland, etc.), there is a high probability of premature birth, and the severity of immunological incompatibility between the mother and the fetus significantly affects the gestation period at which the birth ends [106, 115]. In a number of studies, opinions have been expressed about the allergic reaction of the mother to fetal tissues during pregnancy [116, 117]. According to relevant sources, the amount of eosinophilic granulocytes in the amniotic fluid increases in cases of premature birth. As well, the number of mast cells, more sensitive to allergic processes, is greater in the uterine muscle, and due to the effect of prostaglandin, they easily degranulate and accelerate the accumulation of the uterus. Currently, scientific studies are being conducted to study the effectiveness of antihistamine drugs in preventing premature birth [118].

Congenital anomalies of the uterus, polyhydramnios, and the high risk of premature birth during multiple pregnancy indicate that uterine muscle tension plays an important role in pregnancy disruption. Although the size of the uterus increases as the pregnancy period increases, the intrauterine tension remains constant. Progesterone and endogenous myometrial relaxants, especially nitric oxide, are important in maintaining this stability [119, 120]. Prostaglandins and neuromuscular junction proteins, especially connexin-43, increase during uterine strain [121]. Amniochorial strain, on the other hand, causes the premature rupture of membranes and premature birth. Because of this, premature birth is more difficult to control and is regarded as an almost unavoidable process.

Thus, although numerous concepts of the causes and pathophysiology of premature birth have been put forward and various developmental mechanisms have been determined, it is not possible to completely prevent perinatal pathologies caused by premature termination of pregnancy or incomplete birth. Although inflammation has been identified as the main pathogenetic mechanism in the occurrence of premature birth, the pathophysiology of the process has not yet been fully studied. The inflammatory process in the fetoplacental system is stimulated both by the effect of bacterial colonization and by the effect of hypoxia, accompanied by an inflammatory response in the maternal and fetal bodies. However, since the response to fetal infection is often not manifested by noticeable pathological changes in a pregnant woman, it can be overlooked by specialists or identified late. Even if it does not cause a pathological process in the mother’s body, the fetal inflammatory response causes premature birth, increased perinatal morbidity, and mortality. In modern times, despite the improvement of perinatal medical care technologies, methods for the effective prevention of premature birth have not been developed. The specificity and sensitivity of clinical-instrumental examinations and laboratory markers are very low. Therefore, in recent years, numerous scientific studies have been conducted with the aim of finding new sensitive markers of premature birth and identifying women from the risk group by involving pregnant women in screening examinations.

Advertisement

2. Preterm birth and perinatal prognostic indicators of further age pathologies

Due to the high level of technology and quality medical care in the field of neonatology in recent years, noticeable achievements have been made in reducing the mortality rates of small and extremely small premature babies [100, 122]. In many countries, the criteria for live birth have been changed and great achievements have been made in feeding children whose gestational age is older than 22 weeks [123, 124, 125]. Although the number of survivors among children born at 23–24 weeks of gestational age has increased, a number of early and late complications among them have become a serious medical and social problem. While the pathologies of the neonatal period in children born prematurely are mainly related to respiratory, gastrointestinal, neurological, and nutritional problems, the complications of premature birth are manifested in children’s early age, preschool, school, adolescence, and other developmental periods [126, 127, 128].

The lower the gestational age, the more severe are the long-term consequences of a preterm birth. Even in profoundly premature babies without structural changes in the brain, minimal brain dysfunctions are manifested in further development. Deeply premature babies, whose neonatal period is complicated by various pathologies, always require long-term monitoring and rehabilitation [129, 130]. After discharge from neonatal intensive care units, neuro-developmental abnormalities are the most common problem for premature babies. The most common neurodevelopmental anomalies include cerebral palsy, various forms of psychomotor development and behavioral disorders, as well as vision and hearing problems [131, 132, 133, 134, 135].

The timely detection of neurodevelopmental delays and implementation of rehabilitation measures lead to a significant reduction in the severity and complications of later age problems. Therefore, in a number of developed countries, monitoring the development of premature children is carried out at the state level, and multicenter randomized clinical studies are given great importance. The lower the age of gestation and the degree of morphofunctional maturity of the brain, the more common are secondary brain damage in hypoxia-ischemia, sepsis, necrotic enterocolitis, meningitis and other infectious inflammatory processes; nutritional disorders are more common in these children, which manifests itself in various ways at later ages. It shows with developmental problems of degree [136, 137, 138, 139]. In particular, organic damage to the brain—intraventricular hemorrhage, periventricular leukomalacia, and gross psychomotor developmental disorders with ventriculomegaly, including infantile cerebral palsy, are more likely [140, 141].

Radiological examinations have important diagnostic value in the acute period of brain damage and can form certain ideas about future neurodevelopmental problems [122, 142]. However, at present, the results of early ultrasound examinations are hardly used as a prognostic indicator of neuromotor development. According to the studies by Laptook and co-authors, the specificity and sensitivity of abnormal neurosonography results are very low; cerebral palsy is noted in 9.4% of children with a body mass of less than 1000 g and a normal brain ultrasound examination [143, 144]. In the last few decades, the topical diagnosis of brain injuries has started to be performed using magnetic resonance imaging (MRI), and successful results have been achieved in this direction. While significant changes in brain white matter during the acute phase of central nervous system injuries are detected by ultrasound, MRI is a more sensitive imaging modality for identifying smaller brain lesions [145, 146, 147]. Cerebellar lesions, which are difficult to detect by ultrasound, can also be successfully diagnosed by MRI [148]. As a result of this MRI use, monitoring the subsequent development of children with various changes in the brain has led to the discovery of valuable prognostic information [149]. The positive and negative prognostic value of the changes determined as a result of the examination varies depending on the severity of the damage, the gestational age of the child, and the influence of environmental factors affecting developmental delay. For example, among children with white matter damage at the age of five, while the negative prognostic value of a normal MRI for neurodevelopmental retardation is 100%, the positive prognostic value is 75%, excluding cognitive impairment [150, 151, 152].

Considering the presence of numerous factors that can affect cognitive development in later development, it can be considered legitimate that there is no relationship between perinatal white matter damage and cognitive development disorders. Studies have shown that diffuse white matter damage causes neuromotor retardation in preschool and school-aged children [153, 154]. In the research done at Turku University in Finland, it was noted that in premature babies, this examination is carried out not only in the first days after birth but also at the age equivalent to term birth, which leads to the acquisition of honest prognostic information about the development of neuromotor functions in the first five years of life. The essence of this scientific conclusion is that the changes detected as a result of MRI only in adult brain tissue cause neurodevelopmental disorders at later ages [155]. It is believed that the development of the brain mass is proportional to the growth of the head circumference, and the child’s neuro-motor development can be evaluated and predicted based on this indicator. As a result of MRI examination, it is possible to determine the total volume of the brain, and in children born with a very small mass, the volume of the total or individual parts of the brain has a significant impact on the mental, speech, memory, and psychomotor indicators of the child’s further development [156]. Structural damage detected during MRI examination is not always equivalent to brain dysfunction. Thus, psychomotor development may be completely normal in cases where noticeable pathologies are detected [157]. However, in all cases, MRI examination surpasses many development scales and immunohistochemical markers determined on the basis of clinical indicators according to its prognostic value.

Studies on the assessment of early neurological development in premature infants have shown that cerebral palsy and behavioral-developmental pathologies are of greater relevance. Such pathologies are accompanied by abnormal muscle tone and movement disorders, and the clinical course and prognosis depend on the direction in which the motor functions change. Thus, more functional and sensory disorders are observed in hemiplegia and diplegia, while more motor disorders are found in triplegia and quadriplegia [158].

Cerebral palsy is distinguished according to the degree of severity as follows: mild (the child has only mild movement disorders, general muscle activity does not lag behind age norms), moderate (the child can walk with assistive devices, can sit freely), and severe (the child has the ability to move, no assistive devices can be activated) [159]. Of course, along the course of the pathological process, a number of external interventions and the prescription of drugs significantly affect the prognostic indicators. For example, the administration of antenatal steroids and indomethacin in the perinatal period reduces the risk of intraventricular hemorrhage [160]. Corticosteroids used postnatally in the treatment and prevention of bronchopulmonary dysplasia in premature infants increase the risk of developing cerebral palsy [161, 162, 163]. In general, according to the incidence of psychomotor developmental pathologies of early age, those born with deep prematurity and extreme small mass are at greater risk, with such pathologies occurring in 9–17% of children who survive various perinatal pathologies [164, 165]. Numerous scientific research projects have been carried out on the further development of children born prematurely; over time, the results found through this research have differed fundamentally from one study to another. Information about classical randomized studies entered the periodical literature in the 1970s. If the first epidemiological studies were devoted only to the study of the form and frequency of developmental delays, since the 1990s, the effectiveness of various scales and diagnostic markers to assess psychomotor and sensory development became the objects of research. Within the framework of the International Institute on Child Health and Development, the assessment of children aged 18–22 months after birth began for the first time, using the Classification System of Gross Motor Development, the Bailey Scale of Children’s Development Assessment, and the Amiel–Thison scale of neurological development [92, 150].

In the twentieth century, starting from the perinatal period, various assessment and development programs were implemented one after the other, and large-scale scientific research was carried out in the search for more sensitive indicators of various neuromotor disorders. At present, the Bailey-2 scale of developmental assessment, Denver developmental screening test, CAT/CLAMS scale, and Gessel and Mullen comprehension tests are widely used [134, 166, 167]. The Denver screening test and its modified variants detect and predict gross motor, fine motor, social adaptation, and speech problems in the first 6 years of life starting from the first months after birth [168, 169]. Evaluation by this scale is currently considered the ideal test system for identifying children with developmental delays, playing the role of screening. Evaluation with the Beley scale and its modified versions allows for the detection of the severity of the pathological process in several ways and various clinical forms in children with developmental delays detected through screening [170, 171].

Developmental assessment tests are used to identify and predict the retardation of children’s mental and psychomotor development levels in each developmental period. Based on the obtained results, timely preventive measures can lead to the prevention of a number of developmental delays. However, assessment and prediction based on these scales have several difficulties. Specifically, the accuracy of predictions is influenced by a number of external factors, including the social environment surrounding the child, educational level of the parents, and structural changes of the brain with various origins. Furthermore, it is not always possible to involve parents and children in such assessments in a timely and regular manner given that examinations take a long time.

Cognitive and behavioral reactions are of great importance in the proper formation of children’s social adaptation. Profoundly preterm infants and children with intrauterine growth retardation constitute a greater risk group for the formation of cognitive functions on a weak basis [127, 172]. It is believed that both the antenatal and postnatal retardation of the child lead to a delay in behavioral and cognitive functions [173]. The psychological state of parents has a significant impact on the behavioral problems of premature children. Studies have found that there is a statistically significant relationship between parents’ stress index and children’s behavioral responses at 3 years of age [174]. In another study, it was shown that depressive symptoms in mothers led to impaired social adaptation and behavioral responses of children aged 5 years [175]. Experts who study the impact of nutrition on development have sought to prove that the formation of cognitive reactions does not differ in children who lag behind in antenatal or postnatal development and depends more on proper and balanced nutrition [176, 177]. Many authors have confirmed that there is a dependence between a child’s weight and height parameters at birth and physical, psychomotor, and neurological development at later ages. According to Tanabe K. and co-authors, 18.2% of children with intrauterine growth retardation are stunted at school age. In children born with prematurity and intrauterine growth retardation syndrome, various changes occur in the central nervous system, and many are maintained for a long time without recovery at a later age [39, 119]. Especially in deep premature babies, pathologies such as cerebral hemorrhage, periventricular leukomalacia, and hypertension-hydrocephalus syndrome cannot be fully recovered despite continuous dispensary control, proper intensive therapy, and rehabilitation treatment [87, 178]. P. Casolini created experimental subneurotoxic anoxia in mice in the first week of life and observed persistent dysfunction of the hippocampus and cerebral cortex as well as the disruption of behavioral responses in subsequent development [179]. In another scientific study, the results of magnetic resonance examinations showed that in children born with low weight, the development of various structures of the brain was not proportional—the amount of gray matter in some areas was low and was maintained for a long time [180]. The disproportionate development of brain structures in children born with low weight was accompanied by physical and sexual underdevelopment at different ages, disruption of social adaptation, and attention disorders [171, 181, 182].

Research conducted over many years has concluded that in addition to psychomotor developmental delays, somatic and infectious pathologies of various organs and systems are also more likely to occur in the further development of children born prematurely. Children born with low birth weight have a high risk of death due to cardiovascular pathology [1]. Epidemiological studies conducted jointly by several perinatal scientific centers have proven that intrauterine growth retardation created experimentally as a result of hypoxia and nutrient deficiency in pregnant mice was a major risk factor for the development of cardiovascular diseases in later development is a risk [166, 183, 184]. Left ventricular remodeling of the heart has proven that ultrastructural changes caused by both ischemia and nutrient deficiency lead to future cardiac dysfunction [1].

Research on the genetic basis of organ dysfunction has led to certain results. For example, in studies conducted on mice, the removal of the NR2B fraction from the C terminus of the NMDA receptor gene, which plays an important role in the pathogenesis of perinatal damage to the nervous system, resulted in perinatal death. Another group of researchers observed a substantial decrease in NR1 and NR2B subunits as a result of biochemical tests in mice whose mothers were stressed during pregnancy. Later, electrophysiological studies revealed that intrauterine stress causes memory and cognitive impairments in later life as a result of the long-term dysfunction of the hippocampus [179, 180].

Many diagnostic and preventive programs have been proposed to prevent near and far consequences of preterm birth [130, 185, 186]. A number of sources have suggested a control program for children born with intrauterine growth retardation in polyclinic conditions, which serves to reduce their morbidity [23]. Depending on the variant of intrauterine growth retardation syndrome, dynamic dispensary control leads to a decrease in morbidity in such children. G.K. Swamy, with his research, has shown that timely and correct perinatal care has the leading role in reducing the morbidity and mortality rates of premature babies at a later age [187]. However, he has noted that even state-of-the-art medical care does little to reduce perinatal and postnatal morbidity in children born with severe and extreme low birth weight.

Advertisement

3. Conclusion

Thus, since the mechanisms of the formation of perinatal-period pathologies in children born prematurely have not yet been fully studied, early and late prevention of most diseases associated with the period of intrauterine development is not possible. However, problems of intrauterine development lie in the genesis of many diseases that appear in later development. Endothelial function is the basis of the formation mechanisms of the immune response to the inflammatory process in the fetoplacental system. Not only in the intrauterine period, but also in the early adaptation period after birth, due to the influence of various exogenous and endogenous factors, changes in the vasoregulatory and inflammatory response of the vascular endothelium affect the manifestation of diseases that appear in later development to one degree or another. Given the nature of the relationship between the inflammatory markers of individual organs and systems and the indicators of endothelial dysfunction, it is not possible to determine the perinatal risk factors affecting the future development of the body, but a wide range of scientific research is being conducted in this direction. Investigating the influence of the level of perinatal organ and endothelial dysfunction markers on further development, as well as the regional characteristics of premature birth, can be the basis for creating a perinatal prevention program for early-age pathologies.

References

  1. 1. Gaillard R, Steegers E, Tiemeier H, Hofman A, Jaddoe V. Placental vascular dysfunction, fetal and childhood growth, and cardiovascular development. Circulation. 2013;128:2202-2210
  2. 2. Liu L, Johnson HL, Cousens S, Perin J, Scott S, Lawn JE, et al. Global, regional, and national causes of child mortality: An updated systematic analysis for 2010 with time trends since 2000. Lancet. 2012;379:2151-2161
  3. 3. Katz J, Lee AC, Kozuki N, Lawn JE, Cousens S, et al. Mortality risk in preterm and small-for-gestational-age infants in low-income and middle-income countries: A pooled country analysis. The Lancet. 2013;382:417-425
  4. 4. Risnes KR, Vatten LJ, Baker JL, Jameson K, Sovio U, et al. Birthweight and mortality in adulthood: A systematic review and meta-analysis. International Journal of Epidemiology. 2011;40:647-661
  5. 5. Simchen MJ, Beiner ME, Strauss-Liviathan N, Dulitzky M, Kuint J, et al. Neonatal outcome in growth-restricted versus appropriately grown preterm infants. American Journal of Perinatology. 2000;17:187-192
  6. 6. Souza JP, Gulmezoglu AM, Vogel J, Carroli G, Lumbiganon P, et al. Moving beyond essential interventions for reduction of maternal mortality (the WHO Multicountry Survey on Maternal and Newborn Health): A cross-sectional study. Lancet. 2013;381:1747-1755
  7. 7. Eric CE, Stark AR. Management and outcomes of very low birth weight. The New England Journal of Medicine. 2008;358(16):1701-1711
  8. 8. Trindade CEP. Minerals in the nutrition of extremely low birth weight infants. Jornal de Pediatria. 2005;81(1(Suppl)):43-51
  9. 9. Darlow BA, Cust AE, Donoghue DA. Improved outcomes for very low birth weight infants: Evidence from New Zealand national population based data. Archives of Disease in Childhood. Fetal and Neonatal Edition. 2003;88(1):F23-F28
  10. 10. Koo WW, Hockman EM. Posthospital discharge feeding for preterm infants: Effects of standard compared with enriched milk formula on growth, bone mass, and body composition. American Journal of Clinical Nutrition. 2006;84:1357-1364
  11. 11. Meinzen-Derr J et al. Role of human milk in extremely low birth weight infants' risk of necrotizing enterocolitis or death. Journal of Perinatology. 2009;29:57-62
  12. 12. Carbajal R, Rousset A, Danan C, et al. Epidemiology and treatment of painful procedures in neonates in intensive care units. Journal of the American Medical Association. 2008;300(1):60-70
  13. 13. Grunau RE, Haley DW, Whitfield MF, et al. Altered basal cortisol levels at 3, 6, 8 and 18 months in preterm infants born at extremely low gestational age. The Journal of Pediatrics. 2007;150:151-156
  14. 14. Reddy UM, Ko CW, Raju TNK, Willinger M. Delivery indications at late-preterm gestations and infant mortality rates in the United States. Pediatrics. 2009;124:234-240
  15. 15. Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes of preterm birth. The Lancet. 2008;371(9606):75-84
  16. 16. Mathews TJ, MacDorman MF. Infant mortality statistics from the 2006 period linked birth/infant death data set. National Vital Statistics Reports. 2010;58(17):1-32
  17. 17. Lucja B, Gold C. Early intervention for premature infants in neonatal intensive care unit. Acta Neuropsychologica. 2014;12(2):185-203
  18. 18. Norman J, Greer I. Preterm Labour, Managing Risk in Clinical Practice. Cambridge: Cambridge University Press; 2011
  19. 19. Wallace ME, Mendola P, Chen Z, Grantz KL. Preterm birth in the context of increasing income inequality. Maternal and Child Health Journal. 2016;20(1):164-171. DOI: 10.1007/s10995-015-1816-9
  20. 20. Althabe F, Belizan JM, McClure EM, HemingwayFoday J, Berrueta M, Mazzoni A, et al. A population-based, multifaceted strategy to implement antenatal corticosteroid treatment versus standard care for the reduction of neonatal mortality due to preterm birth in low-income and middle-income countries: The ACT cluster-randomised trial. Lancet. 2015;385(9968):629-639
  21. 21. Conde-Agudelo A, Diaz-Rossello JL. Kangaroo mother care to reduce morbidity and mortality in low birthweight infants. Cochrane Database of Systematic Reviews. 2014;4:CD002771
  22. 22. Kinney MV, Lawn JE, Howson CP, Belizan J. 15 million preterm births annually: What has changed this year? Reproductive Health. 2012;9:28
  23. 23. Black RE, Cousens S, Johnson HL, Lawn JE, Rudan I, Bassani DG, et al. Global, regional, and national causes of child mortality in 2008: A systematic analysis. Lancet. 2010;375(9730):1969-1987
  24. 24. Wong L, Wilkes J, Korgenski K, Varner MW, Manuck TA. Risk factors associated with preterm birth after a prior term delivery. BJOG. 2016;123(11):1772-1778. DOI: 10.1111/1471-0528.13683
  25. 25. Norwitz ER, Phaneuf LE, Caughey AB. Progesterone supplementation and the prevention of preterm birth. Reviews in Obstetrics and Gynecology. 2011;4(2):60-72
  26. 26. McDonald SD, Han Z, Mulla S, Murphy KE, Beyene J, Ohlsson A. Preterm birth and low birth weight among in vitrofertilization singletons: A systematic review and meta-analyses. EJOG. 2009;146(2):138-148
  27. 27. Wisborg K, Ingerslev HJ, Henriksen TB. In vitro fertilization and preterm delivery, low birth weight, and admission to the neonatal intensive care unit: A prospective follow-up study. Fertility and Sterility. 2010;94(6):2102-2106
  28. 28. Lu L, Qu Y, Tang J, Chen D, Mu D. Risk factors associated with late preterm births in the underdeveloped region of China: A cohort study and systematic review. Taiwanese Journal of Obstetrics and Gynecology. 2015;54:647-653
  29. 29. Barber MA, Eguiluz I, Plasencia W, Medina M, Valle L, Garcia JA. Preterm delivery and ultrasound measurement of cervical length in Gran Canaria, Spain. International Journal of Gynecology & Obstetrics. 2010;108:58-60
  30. 30. Mesiano S, Wang X, Norwitz ER. Progesterone receptors in the human pregnancy uterus: Do they hold the key to birth timing? Reproductive Sciences. 2011;18:6-19
  31. 31. Asuquo B, Vellone AD, Walters G, Manney S, Miquimi L, Kurst H. A case-control study of teh risk of adverse perinatal outcomes due to tuberculosis during pregnancy. Journal of Obstetrics and Gynaecology. 2012;32:635-638. DOI: 10.3109/01443615. 2012.704436
  32. 32. George LM, Nadeem OK, Quinlivan JA. Bacterial aetiological agents of intra-amniotic infections and preterm birth in pregnant women. Frontiers in Cellular and Infection Microbiology. 2013;3:58-65
  33. 33. Hobel CJ. Stress and preterm birth. Clinical Obstetrics and Gynecology. 2004;47(4):856-880
  34. 34. McDonald SW, Kingston D, Bayrampour H, Dolan SM, Tough SC. Cumulative psychosocial stress, coping resources, and preterm birth. Archives of Women's Mental Health. 2014;17(6):559-568. DOI: 10.1007/s00737-014-0436-5
  35. 35. Berit D, Pal R, Pal O, Lars J, Anne E. Placenta weight in pre-eclampsia. Acta Obstetricia et Gynecologica Scandinavica. 2008;87(6):608-611
  36. 36. Lacobellia S, Bonsantea F, Robillard P. Pre-eclampsia and preterm birth in Reunion island: A 13 years cohort based study. Comparison with international data. Journal of Maternal-Fetal and Neonatal Medicine. 2016;29(18):3035-3040. DOI: 10.3109/14767058.2015.1114081
  37. 37. Royal College of Obstetricians and Gynaecologists. The Management of Severe Pre-eclampsia/Eclampsia; National Institute for Health and Clinical Excellence (2008) Antenatal Care. NICE Clinical Guideline 62. London: National Institute for Health and Clinical Excellence; 2006
  38. 38. Brian MM. Preterm premature rupture of the membranes: Diagnosis and management. Clinics in Perinatology. 2004;31(4):765-782
  39. 39. Ahmed A, Rahman M, Zhang X, Acevedo CH, Nijjar S, Rushton I, et al. Induction of placental heme oxygenase-1 is protective against TNFalpha-induced cytotoxicity promotes vessel relaxation. Molecular Medicine. 2000;6:391-409
  40. 40. George EM, Stout JM, Stec DE, Granger JP. Heme oxygenase induction attenuates TNF-α-induced hypertension in pregnant rodents. Frontiers in Pharmacology. 2015;6:165. DOI: 10.3389/fphar.2015.00165
  41. 41. Kalantaridou SN, Zoumakis E, Makrigiannakis A, Lavasidis LG, Vrekoussis T, Chrousos GP. Corticotropin-releasing hormone, stress and humanreproduction: An update. Journal of Reproductive Immunology. 2010;85:33-39
  42. 42. Wadhwa PD, Garite T, Porto M, Sandman CA. Corticotropin-releasing hormone (CRH), preterm birth and fetal growth restriction: A prospective investigation. American Journal of Obstetrics and Gynecology. 2004;191(4):1063-1069
  43. 43. Keller PA, Kirkwood K, Morgan J, Westcott S, McCluskey A. The prevention of preterm labour—Corticotropin releasing hormone type 1 receptors as a target for drug design and development. Mini-Reviews in Medicinal Chemistry. 2016;3(4):295-303
  44. 44. Torricelli M, Petraglia F. Placental hormones and identification of pregnancy at risk. Gynecological Endocrinology. 2010;26:705-707
  45. 45. Voltolini C, Petraglia F. Neuroendocrinology of pregnancy and parturition. Clinical Neuroendocrinology. 2014;124:17-36. DOI: 10.1016/B978-0-444-59602-4.00002-2.
  46. 46. Berghella V, Figueroa D, Szychowski JM, et al. 17-alpha-hydroxyprogesterone caproate for the prevention of preterm birth in women with prior preterm birth and a short cervical length. American Journal of Obstetrics and Gynecology. 2010;202(4):351, e1-6
  47. 47. Committee on Practice Bulletins-The American College of Obstetricians and Gynecologists. Practice bulletin no. 130: Prediction and prevention of preterm birth. Obstetrics and Gynecology. 2012;120(4):964-973
  48. 48. Dodd JM, Jones L, Flenady V, Cincotta R, Crowther CA. Prenatal administration of progesterone for preventing preterm birth in women considered to be at risk of preterm birth. Cochrane Database of Systematic Reviews. 2013;7:CD004947. DOI: 10.1002/14651858
  49. 49. Tita ATN, Andrews WW. Diagnosis and management of clinical chorioamnionitis. Clinics in Perinatology. 2010;37(2):339-354
  50. 50. Kadhim HJ, Duchateau J, Sebire G. Cytokines and brain injury: Invited review. Journal of Intensive Care Medicine. 2008;23:236-249
  51. 51. Dammann O, Leviton A. Inflammatory brain damage in preterm newborns—Dry numbers, wet lab, and causal inferences. Early Human Development. 2004;79:1-15
  52. 52. Malaeb S, Dammann O. Fetal inflammatory response and brain injury in the preterm newborn. Journal of Child Neurology. 2009;24(9):1119-1126
  53. 53. Sciaky-Tamir Y, Hershkovitz R, Mazor M, Shelef I, Erez O. The use of imaging technology in the assessment of the fetal inflammatory response syndrome—Imaging of the fetal thymus. Prenatal Diagnosis. 2015;35(5):413-419
  54. 54. Viscardi RM. Perinatal inflammation and lung injury. Seminars in Fetal & Neonatal Medicine. 2012;17(1):30-35
  55. 55. Hoang M, Potter JA, Gysler SM, Han CS, Guller S, Norwitz ER, et al. Human fetal membranes generate distinct cytokine profiles in response to bacterial Toll-like receptor and nod-like receptor agonists. Biology of Reproduction. 2014;90(2):39
  56. 56. Paananen R, Husa AK, Vuolteenaho R, Herva R, Kaukola T, Hallman M. Blood cytokines during the perinatal period in very preterm infants: Relationship of inflammatory response and bronchopulmonary dysplasia. The Journal of Pediatrics. 2009;154(39-43):e33
  57. 57. Adams W, Rubens CE, Gravett MG. Use of nonhuman primate models to investigate mechanisms of infection-associated preterm birth. BJOG. 2011;118:136-144
  58. 58. DiGiulio DB, Gervasi MT, Romero R, Vaisbuch E, Mazaki-Tovi S, Kusanovic JP, et al. Microbial invasion of the amniotic cavity in pregnancies with small-for-gestational-age fetuses. Journal of Perinatal Medicine. 2010a;38:495-502
  59. 59. Bowes WA. The role of antibiotics in the prevention of preterm birth. F1000 Medicine Reports. 2009;1:22
  60. 60. Bookstaver PB, Bland CM, Griffin B, Stover KR, Eiland LS, McLaughlin M. A review of antibiotic use in pregnancy. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy. 2015;35(11):1052-1062
  61. 61. Smaill F, Vazquez JC. Antibiotics for Asymptomatic Bacteriuria in Pregnancy (Review). Cochrane Database Syst Rev. 7 Aug 2015;(8):CD000490. DOI: 10.1002/14651858.CD000490.pub3. Update in: Cochrane Database Syst Rev. 25 Nov 2019;2019(11). PMID: 26252501.
  62. 62. Thinkhamrop J, Hofmeyr GJ, Adetoro O, Lumbiganon P. Prophylactic antibiotic administration in pregnancy to prevent infectious morbidity and mortality. Cochrane database of systematic reviews (online). 2002;4(4):cd002250
  63. 63. Locksmith G, Duff P. Infection, antibiotics, and preterm delivery. Seminars in Perinatology. 2001;25(5):295-309
  64. 64. Morency A, Bujold E. The effect of second-trimester antibiotic therapy on the rate of preterm birth. JOGC. 2007;29(1):35-44
  65. 65. Simcox R, Sin WT, Seed PT, Briley A, Shennan AH. Prophylactic antibiotics for the prevention of preterm birth in women at risk: A meta-analysis. The Australian & New Zealand Journal of Obstetrics & Gynaecology. 2007;47:368-377
  66. 66. Farag AH, Mohammed MM, Ellaithy MI, Salama HA. Blind vaginal fetal fibronectin swab for prediction of preterm birth. Journal of Obstetrics and Gynaecology Research. 2015;41(7):1009-1017
  67. 67. Kiefer DG, Vintzileos AM. The utility of Fetal fibronectin in the prediction and prevention of spontaneous preterm birth. Reviews in Obstetrics and Gynecology. 2008;1(3):106-112
  68. 68. Brian MM, Yolanda AR, Gary RT, Menachem MR, Robert LG, Anita FD, et al. The NICHD-MFMU antibiotic treatment of preterm PROM study: Impact of initial amniotic fluid volume on pregnancy outcome. American Journal of Obstetrics and Gynecology. 2006;194(2):438-445
  69. 69. Joy S. Nichd Mfmu Network. Latency and infectious complications following preterm premature rupture of the membranes: Impact of body mass index. American Journal of Obstetrics and Gynecology. 2009;201(6):600.e1-5. DOI: 10.1016/j.ajog.2009.06.030
  70. 70. Mercer BM, Crouse DT, Goldenberg RL, Miodovnik M, Mapp DC, Meis PJ, et al. The antibiotic treatment of PPROM study: Systemic maternal and fetal markers and perinatal outcomes. American Journal of Obstetrics and Gynecology. 2012;206(2):145.e1-145.e9
  71. 71. Harirah H, Donita SE, Hsu CD. Amniotic fluid matrix metalloproteinase-9 and interleukin-6 in predicting intra-amniotic infection. Obstetrics and Gynecology. 2002;99:80-85
  72. 72. Lee SE, Romero R, Jung H, Park CW, Park JS, Yoon BH. The intensity of the fetal inflammatory response in intraamniotic inflammation with and without microbial invasion of the amniotic cavity. American Journal of Obstetrics and Gynecology. 2007;197(294):e1-294.e6
  73. 73. Taylor BD, Holzman CB, Fichorova RN, Tian Y, Jones NM, Wenjiang F, et al. Inflammation biomarkers in vaginal fluid and preterm delivery. Human Reproduction. 2013;28(4):942-952
  74. 74. O’Brien CM, Arbuckle S, Thomas S, Rode J, Turner R, Jeffery HE. Placental inflammation is associated with rural and remote residence in the Northern Territory, Australia: A cross-sectional study. BMC Pregnancy and Childbirth. 2015;15:32
  75. 75. Lee SE, Romero R, Park CW, Jun JK, Yoon BH. The frequency and significance of intraamniotic inflammation in patients with cervical insufficiency. American Journal of Obstetrics and Gynecology. 2008;198:633.e1-633.e8
  76. 76. Queenan JT, Spong CY, Lockwood CJ. Protocols for High-Risk Pregnancies: An Evidence-Based Approach. Wiley-Blackwell. Available from: https://doi.org/10.1002/9781444323870
  77. 77. Suhag A, Berghella V. Cervical cerclage. Clinical Obstetrics and Gynecology. 2014;57(3):557-567
  78. 78. Kristen L, Camilo A, Clinton J, Nelson M, Brian M, Cameron MJ, et al. Differential receptor for advanced glycation end products expression in preeclamptic, intrauterine growth restricted, and gestational diabetic placentas. American Journal of Reproductive Immunology. 2016;75(2):172-180
  79. 79. Rzepka R, Dołegowska B, Rajewska A, Kwiatkowski S, Sałata D, Budkowska M, et al. Soluble and endogenous secretory receptors for advanced glycation end products in threatened preterm labor and preterm premature rupture of fetal membranes. BioMed Research International. 2015;2015; Article ID 568042, p. 10
  80. 80. Schaefer TM, Desouza K, Fahey JV, Beagley KW, Wira CR. Toll-like receptor (TLR) expression and TLR-mediated cytokine/chemokine production by human uterine epithelial cells. Immunology. 2004;112(3):428-436
  81. 81. Buhimschi IA, Zhao G, Pettker CM. The receptor for advanced glycation end products (RAGE) system in women with intraamniotic infection and inflammation. American Journal of Obstetrics and Gynecology. 2007;196(181):e1-181.e12
  82. 82. Dubicke A, Andersson P, Fransson E, Andersson E, Sioutas A, et al. High-mobility group box protein 1 and its signalling receptors in human preterm and term cervix. Journal of Reproductive Immunology. 2010;84(1):86-94
  83. 83. Thaxton JE, Nevers TA, Sharma S. TLR-mediated preterm birth in response to pathogenic agents. Infectious Diseases in Obstetrics and Gynecology. 2010;2010, pii: 378472
  84. 84. Noguchi T, Sado T, Naruse K, Shigetomi H, Onogi A, Haruta S, et al. Evidence for activation of Toll-like receptor and receptor for advanced glycation end products in preterm birth. Mediators of Inflammation. 2010:1-10. Article ID 490406
  85. 85. Bastek JA, Brown AG, Foreman MN, McShea MA, Anglim LM, Adamczak JE, et al. The soluble receptor for advanced glycation end products can prospectively identify patients at greatest risk for preterm birth. The Journal of Maternal-Fetal & Neonatal Medicine. 2012;25:1762-1768
  86. 86. Romero R, Chaiworapongsa T, Savasan ZA, Hussein Y, Dong Z, Kusanovic JP, et al. Clinical chorioamnionitis is characterized by changes in the expression of the alarmin HMGB1 and one of its receptors, sRAGE. The Journal of Maternal-Fetal & Neonatal Medicine. 2012;25:558-567
  87. 87. Bezold KY, Karjalainen MK, Hallman M, Teramo K, Muglia LJ. The genomics of preterm birth: From animal models to human studies. Genome Medicine. 2013;5:34
  88. 88. Krediet TG, Wiertsema SP, Vossers MJ, Hoeks SBEA, Fleer A, Ruven HJT, et al. Toll-like receptor 2 polymorphism is associated with preterm birth. Pediatric Research. 2007;62(4):474-476
  89. 89. Espinoza J. Uteroplacental ischemia in early- and late-onset pre-eclampsia: A role for the fetus? Ultrasound in Obstetrics & Gynecology. 2012;40(4):373-382
  90. 90. Gilbert JS, Bauer AJ, Gingery A, Chasson S. Circulating and utero-placental adaptations to chronic placental ischemia in the rat. Placenta. 2011;33(2):100-105
  91. 91. Granger JP, Alexander BT, Llinas MT, Bennett WA, Khalil RA. Pathophysiology of preeclampsia: Linking placental ischemia/hypoxia with microvascular dysfunction. Microcirculation. 2002;9(3):147-160
  92. 92. Thompson RJ, Goldstein RF, Oehler JM, Gustafson KE, Catlett AT, Brazy JE. Developmental outcome of very low birth weight infants as a function of biological risk and psychosocial risk. Journal of Developmental and Behavioral Pediatrics. 1994;15(4):232-238
  93. 93. Van Iersse SH, Conraads VM, Van Craenenbroeck EM, Liu Y, Maas A, Parizel PM, et al. Endothelial dysfunction in acute brain injury and the development of cerebral ischemia. Journal of Neuroscience Research. 2015;93(6):866-872
  94. 94. Livrinova V, Hadzi Lega M, Hristova Dimcheva A, Samardziski I, Isjanovska R. Factor V Leiden, prothrombin and MTHFR mutation in patients with Preeclamsia, intrauterine growth restriction and placental abruption. Open Access Macedonian Journal of Medical Sciences. DOI: 10.3889/oamjms.2015.099
  95. 95. Chighizola CB, Andreoli L, de Jesus GR, Banzato A, Pons-Estel GJ, Erkan D. The association between antiphospholipid antibodies and pregnancy morbidity, stroke, myocardial infarction, and deep vein thrombosis: A critical review of the literature. Lupus. 2015;24:980-984
  96. 96. Giancotti A, La Torre R, Spagnuolo A, D’Ambrosio V, Cerekja A, Piazze J. Efficacy of three different antithrombotic regimens on pregnancy outcome in pregnant women affected by recurrent pregnancy loss. The Journal of Maternal-Fetal & Neonatal Medicine. 2012;25(7):1191-1194
  97. 97. Karakantza M, Androutsopoulos G, Mougiou A, Decavalas GO. Inheritance and perinatal consequences of inherited thrombophilia in Greece. International Journal of Gynecology & Obstetrics. 2008;100(2):124-129
  98. 98. Dhobale M, Chavan P, Kulkarni A, Mehendale S, Pisal H, Joshi S. Reduced folate, increased vitamin B(12) and homocysteine concentrations in women delivering preterm. Annals of Nutrition and Metabolism. 2012;61:7-14
  99. 99. Bergen NE, Jaddoe VWV, Timmermans S, et al. Homocysteine and folate concentrations in early pregnancy and the risk of adverse pregnancy outcomes: The generation R study. British Journal of Obstetrics and Gynaecology. 2012;119(6):739-751
  100. 100. Blencowe H, Cousens S, Oestergaard M, Chou D, Moller AB, Narwal R, et al. National, regional and worldwide estimates of preterm birth. The Lancet. 2012;379(9832):2162-2172
  101. 101. Charles DHM, Ness AR, Campbell D, Smith GD, Whitley E, Hall MH. Folic acid supplements in pregnancy and birth outcome: Re-analysis of a large randomised controlled trial and update of Cochrane review. Paediatric and Perinatal Epidemiology. 2005;19(2):112-124
  102. 102. Lee J, Romero R, Xu Y, Kim JS, Topping V, Yoo W, et al. A signature of maternal anti-fetal rejection in spontaneous preterm birth: Chronic chorioamnionitis, anti-human leukocyte antigen antibodies, and C4d. PLoS One. 2011;6(2):e16806
  103. 103. Young RC. Synchronization of regional contractions of human labor; direct effects of region size and tissue excitability. Journal of Biomechanics. 2015;48(9):1614-1619
  104. 104. Lucovnik M, Kuon RJ, Chambliss LR, Maner WL, Shi S, Leili Shi L, et al. Progestin treatment for the prevention of preterm birth. Acta Obstetricia et Gynecologica Scandinavica. 2011;90(10):1057-1069
  105. 105. Romero R, Yeo L, Chaemsaithong P, Chaiworapongsa T, Hassan SS. Progesterone to prevent spontaneous preterm birth. Seminars in Fetal & Neonatal Medicine. 2014;19(1):15-26
  106. 106. Zhang H, Zhang L. Role of protein kinase C isozymes in the regulation of alpha1-adrenergic receptor-mediated contractions in ovine uterine arteries. Biology of Reproduction. 2008;78(1):35-42
  107. 107. Xiao D, Longo LD, Zhang L. Alpha1-adrenoceptor-mediated phosphorylation of MYPT-1 and CPI-17 in the uterine artery: Role of ERK/PKC. American Journal of Physiology. Heart and Circulatory Physiology. 2005;288(6):H2828-H2835
  108. 108. An BS, Ahn HJ, Kang HS, Jung EM, Yang H, Hong EJ, et al. Effects of estrogen and estrogenic compounds, 4-tert-octylphenol, and bisphenol A on the uterine contraction and contraction-associated proteins in rats. Molecular and Cellular Endocrinology. 2013;375(1-2):27-34
  109. 109. Elmes M, Szyszka A, Pauliat C, Clifford B, Daniel Z, Cheng Z, et al. Maternal age effects on myometrial expression of contractile proteins, uterine gene expression, and contractile activity during labor in the rat. Physiological Reports. 2015;3(4):e12305
  110. 110. Cook JL, Zaragoza DB, Sung DH, Olson DM. Expression of myometrial activation and stimulation genes in a mouse model of preterm labor: Myometrial activation, stimulation, and preterm labor. Endocrinology. 2000;141(5):1718-1728
  111. 111. Chinnathambi V, Blesson CS, Vincent KL, Saade GR, Hankins GD, Yallampalli C, et al. Elevated testosterone levels during rat pregnancy cause hypersensitivity to angiotensin II and attenuation of endothelium-dependent vasodilation in uterine arteries. Hypertension. 2014;64:405-414
  112. 112. Gokina NI, Kuzina OY, Vance AM. Augmented EDHF signaling in rat uteroplacental vasculature during late pregnancy. American Journal of Physiology. Heart and Circulatory Physiology. 2010;299:H1642-H1652
  113. 113. Lee J, Romero R, Xu Y, Kim JS, Park JY, Kusanovic JP, et al. Maternal HLA panel-reactive antibodies in early gestation positively correlate with chronic chorioamnionitis: Evidence in support of the chronic nature of maternal anti-fetal rejection. American Journal of Reproductive Immunology. 2011;66(6):510-526
  114. 114. Tirado-González I, Nancy Freitag N, Gabriela Barrientos G, Blois SM. Galectin-I influences trophoblast immune evasion and emerges as a predictive factor for the outcome of pregnancy. Molecular Human Reproduction. 2013;19(1):43-53
  115. 115. Khashan AS, Kenny LS, Laursen TM, Mahmood U, Mortensen PB, Henriksen TB, et al. Pregnancy and the risk of autoimmune disease. PLoS One. 2011;6(5):e19658
  116. 116. Bytautiene E, Romero R, Vedernikov YP. Induction of premature labor and delivery by alergic reaction and prevention by histamin H1 receptor antagonist. American Journal of Obstetrics and Gynecology. 2004;191:1356-1361
  117. 117. Williams Z. Inducing tolerance to pregnancy. New England Journal of Medicine. 2012;367:1159-1161
  118. 118. Kar S, Krishnan A, Preetha K, Mohankar A. A review of antihistamines used during pregnancy. Journal of Pharmacology and Pharmacotherapeutics. 2012;3(2):105-108
  119. 119. Adams Waldorf KM, Singh N, Mohan AR. Uterine overdistention induces preterm labor mediated by inflammation: Observations in pregnant women and nonhuman primates. American Journal of Obstetrics and Gynecology. 2015;213(830):e1-e19
  120. 120. Lee YH, Shynlova O, Lye SJ. Stretchinduced human myometrial cytokines enhance immune cell recruitment via endothelial activation. Cellular & Molecular Immunology. 2015;12:231-242
  121. 121. Davidson JO, Drury PP, Green CR, Nicholson LF, Bennet L, Gunn AJ. Connexin hemichannel blockade is neuroprotective after asphyxia in preterm fetal sheep. PLoS One. 2014;9(5):e96558
  122. 122. Kirton A. Modeling developmental plasticity after perinatal stroke: Defining central therapeutic targets in cerebral palsy. Pediatric Neurology. 2013;48(2):81-94
  123. 123. Sclowitz IK, Santos IS, Domingues MR, Matijasevich A, Barros AJ. Prognostic factors for low birthweight repetition in successive pregnancies: A cohort study. BMC Pregnancy and Childbirth. 2013;13:20. DOI: 10.1186/1471-2393-13-20
  124. 124. Villar J, Giuliani F, Bhutta ZA, Bertino E, Ohuma EO, Ismail LC, et al. Postnatal growth standards for preterm infants: The preterm postnatal follow-up study of the INTERGROWTH-21(st) project. The Lancet Global Health. 2015;3(11):e681-e691
  125. 125. Yee LM, Truong YN, Caughey AB, Cheng YW. The association between interdelivery interval and adverse perinatal outcomes in a diverse US population. Journal of Perinatology. 2016;36(8):593-597. DOI: 10.1038/jp.2016.54
  126. 126. Lefebvre F, Gagnon MM, Luu TM, Lupien G, Dorval V. In extremely preterm infants, do the movement assessment of infants and the Alberta infant motor scale predict 18-month outcomes using the Bayley-III? Early Human Development. 2016;94:13-17. DOI: 10.1016/j.earlhumdev.2016.01.012
  127. 127. Ramel SE, Demerath EW, Gray HL, Younge N, Boys C, Georgieff MK. The relationship of poor linear growth velocity with neonatal illness and two-year neurodevelopment in preterm infants. Neonatology. 2012;102(1):19-24
  128. 128. Rimol LM, Bjuland KJ, Løhaugen GC, Martinussen M, Evensen KA, Indredavik MS, et al. Cortical trajectories during adolescence in preterm born teenagers with very low birthweight. Cortex. 2016;75:120-131. DOI: 10.1016/j.cortex.2015.12.001
  129. 129. Doyle LW, Cheong JL, Burnett A, Roberts G, Lee KJ, Anderson PJ. Victorian infant collaborative study group. Biological and social influences on outcomes of extreme-preterm/low-birth weight adolescents. Pediatrics. 2015;136(6):e1513-e1520
  130. 130. Spittle A, Orton J, Anderson PJ, Boyd R, Doyle LW. Early developmental intervention programmes provided post hospital discharge to prevent motor and cognitive impairment in preterm infants. Cochrane Database of Systematic Reviews. 2015;11:CD005495. DOI: 10.1002/14651858
  131. 131. Coq JO, Delcour M, Massicotte VS, Baud O, Barbe MF. Prenatal ischemia deteriorates white matter, brain organization, and function: Implications for prematurity and cerebral palsy. Developmental Medicine & Child Neurology. 2016;58(4):7-11
  132. 132. Eneriz-Wiemer M, Saynina O, Sundaram V, Lee HC, Bhattacharya J, Sanders LM. Parent language: A predictor for neurodevelopmental follow-up care among infants with very low birth weight. Academic Pediatrics. 2016;16(7):645-52. DOI: 10.1016/j.acap.2016.04.004.
  133. 133. Huseynova SA, Panakhova NF, Hajiyeva AS, Mukhtarova SN, Agayeva GT. Endothelial dysfunction and developmental outcomes of very low birth weight newborns with hypoxic encephalopathy. JPMA. 2017;67(12):1857-1863
  134. 134. Ramoğlu M, Kavuncuoğlu S, Aldemir E, Yarar C, Eras Z. Neurodevelopment of preterms born after IVF and spontaneous multiple pregnancies. Pediatrics International. 2016;58(12)1284-1290. DOI: 10.1111/ped.13012
  135. 135. Szczapa T, Karpiński Ł, Moczko J, Weindling M, Kornacka A, Wróblewska K, et al. Comparison of cerebral tissue oxygenation values in full term and preterm newborns by the simultaneous use of two near-infrared spectroscopy devices: An absolute and a relative trending oximeter. Journal of Biomedical Optics. 2013;18(8):87006. DOI: 10.1117/1.JBO.18.8.087006
  136. 136. Haller S, Deindl P, Cassini A, Suetens C, Zingg W, Abu Sin M, et al. Neurological sequelae of healthcare-associated sepsis in very-low-birthweight infants: Umbrella review and evidence-based outcome tree. Euro Surveillance. 2016;21(8). DOI: 10.2807/1560-7917.ES.2016.21.8.30143
  137. 137. Lee JY, Ahn TG, Jun JK. Short-term and long-term postnatal outcomes of expectant management after previable preterm premature rupture of membranes with and without persistent oligohydramnios. Obstetrics and Gynecology. 2015;126(5):947-953
  138. 138. Milner KM, Neal EF, Roberts G, Steer AC, Duke T. Long-term neurodevelopmental outcome in high-risk newborns in resource-limited settings: A systematic review of the literature. Paediatrics and International Child Health. 2015;35(3):227-242
  139. 139. Miyazaki K, Furuhashi M, Ishikawa K, Tamakoshi K, Hayashi K, Kai A, et al. Impact of chorioamnionitis on short- and long-term outcomes in very low birth weight preterm infants: The Neonatal Research Network Japan. The Journal of Maternal-Fetal & Neonatal Medicine. 2016;29(2):331-337
  140. 140. Radic JA, Vincer M, McNeely PD. Outcomes of intraventricular hemorrhage and posthemorrhagic hydrocephalus in a population-based cohort of very preterm infants born to residents of Nova Scotia from 1993 to 2010. Journal of Neurosurgery. Pediatrics. 2015;15(6):580-588
  141. 141. Wu T, Fan XP, Wang WY, Yuan TM. Enterovirus infections are associated with white matter damage in neonates. Journal of Paediatrics and Child Health. 2014;50(10):817-822
  142. 142. Li D, Hodge J, Wei XC, Kirton A. Reduced ipsilesional cortical volumes in fetal periventricular venous infarction. Stroke. 2012;43(5):1404-1407
  143. 143. Pappas A, Kendrick DE, Shankaran S, Stoll BJ, Bell EF, Laptook AR, et al. Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Chorioamnionitis and early childhood outcomes among extremely low-gestational-age neonates. JAMA Pediatrics. 2014;168(2):137-147
  144. 144. Stoll BJ, Hansen NI, Bell EF, Walsh MC, Carlo WA, Shankaran S, et al. Trends in care practices, morbidity, and mortality of extremely preterm neonates, 1993-2012. Journal of the American Medical Association. 2015;314(10):1039-1051
  145. 145. Jary S, Kmita G, Wroblewska J, Whitelaw A. Quantitative cranial ultrasound prediction of severity of disability in premature infants with post-haemorrhagic ventricular dilatation. Archives of Disease in Childhood. 2012;97:955-959
  146. 146. Ramenghi LA, Fumagalli M, Righini A, Bassi L, Groppo M, Parazzini C, et al. Magnetic resonance imaging assessment of brain maturation in preterm neonates with punctate white matter lesions. Neuroradiology. 2007;49:161-167
  147. 147. Smyser CD, Kidokoro H, Inder TE. Magnetic resonance imaging of the brain at term equivalent age in extremely premature neonates: To scan or not to scan? Journal of Paediatrics and Child Health. 2012;48:794-800
  148. 148. Lee W, Al-Dossary H, Raybaud C, Young JM, Morgan BR, Whyte HE, et al. Longitudinal cerebellar growth following very preterm birth. Journal of Magnetic Resonance Imaging. 2016;43(6):1462-1473. DOI: 10.1002/jmri.25098
  149. 149. Navarra R, Sestieri C, Conte E, Salomone R, Mattei PA, Romani GL, et al. Perinatal MRI diffusivity is related to early assessment of motor performance in preterm neonates. The Neuroradiology Journal. 2016;29(2):137-145
  150. 150. Arichi T, Counsell SJ, Allievi AG, Chew AT, Martinez-Biarge M, Mondi V, et al. The effects of hemorrhagic parenchymal infarction on the establishment of sensori-motor structural and functional connectivity in early infancy. Neuroradiology. 2014;56(11):985-994
  151. 151. Kidokoro H, Anderson PJ, Doyle LW, Woodward LJ, Neil JJ, Inder TE. Brain injury and altered brain growth in preterm infants: Predictors and prognosis. Pediatrics. 2014;134(2):e444-e453
  152. 152. Merchant N, Azzopardi D. Early predictors of outcome in infants treated with hypothermia for hypoxic-ischaemic encephalopathy. Developmental Medicine and Child Neurology. 2015;57(Suppl. 3):8-16
  153. 153. Massaro AN, Evangelou I, Fatemi A, Vezina G, Mccarter R, Glass P, et al. White matter tract integrity and developmental outcome in newborn infants with hypoxic-ischemic encephalopathy treated with hypothermia. Developmental Medicine and Child Neurology. 2015;57(5):441-448
  154. 154. Thompson DK, Chen J, Beare R, Adamson CL, Ellis R, Ahmadzai ZM, et al. Structural connectivity relates to perinatal factors and functional impairment at 7years in children born very preterm. NeuroImage. 2016;134:328-337
  155. 155. Setänen S, Haataja L, Parkkola R, Lehtonen L, Lind A. Predictive value of neonatal brain MRI on the neurodevelopmental outcome of preterm infants by 5 years of age. Acta Paediatrica. 2013;102:492-497
  156. 156. Lind A, Haataja L, Rautava L, Valiaho A, Lehtonen L, Lapinleimu H, et al. Relations between brain volumes, neuropsychological assessment and parental questionnaire in prematurely born children. European Child & Adolescent Psychiatry. 2010;19:407-417
  157. 157. Sølsnes AE, Sripada K, Yendiki A, Bjuland K, Østgård HF, Aanes S, et al. Limited microstructural and connectivity deficits despite subcortical volume reductions in school-aged children born preterm with very low birth weight. NeuroImage. 2016;130:24-34
  158. 158. Rahmati H, Martens H, Aamo OM, Stavdahl Ø, Støen R, Adde L. Frequency-based features for early cerebral palsy prediction. Conference Proceedings: Annual International Conference of the IEEE Engineering in Medicine and Biology Society. 2015;2015:5187-5190. DOI: 10.1109/EMBC.2015.7319560
  159. 159. Strand KM, Dahlseng MO, Lydersen S, Rø TB, Finbråten AK, Jahnsen RB, et al. Growth during infancy and early childhood in children with cerebral palsy: A population-based study. Developmental Medicine and Child Neurology. 2016;58(9):924-30. DOI: 10.1111/dmcn.13098
  160. 160. Verma R, Shibly S, Fang H, Pollack S. Do early postnatal body weight changes contribute to neonatal morbidities in the extremely low birth weight infants. Journal of Neonatal-Perinatal Medicine. 2015;8(2):113-118
  161. 161. Bhatt AJ, Feng Y, Wang J, Famuyide M, Hersey K. Dexamethasone induces apoptosis of progenitor cells in the subventricular zone and dentate gyrus of developing rat brain. Journal of Neuroscience Research. 2013;91(9):1191-1202
  162. 162. Linsell L, Malouf R, Morris J, Kurinczuk JJ, Marlow N. Prognostic factors for cerebral palsy and motor impairment in children born very preterm or very low birthweight: A systematic review. Developmental Medicine and Child Neurology. 2015:169(12):1162-1172. DOI: 10.1111/dmcn.12972
  163. 163. Zia MT, Vinukonda G, Vose LR, Bhimavarapu BB, Iacobas S, Pandey NK, et al. Postnatal glucocorticoid-induced hypomyelination, gliosis, and neurologic deficits are dose-dependent, preparation-specific, and reversible. Experimental Neurology. 2015;263:200-213
  164. 164. Schieve LA, Tian LH, Rankin K, Kogan MD, Yeargin-Allsopp M, Visser S, et al. Population impact of preterm birth and low birth weight on developmental disabilities in US children. Annals of Epidemiology. 2016;26(4):267-274
  165. 165. Slaughter LA, Bonfante-Mejia E, Hintz SR, Dvorchik I, Parikh NA. Early conventional MRI for prediction of neurodevelopmental impairment in extremely-low-birth-weight infants. Neonatology. 2016;110(1):47-54
  166. 166. Bolduc ME, Du Plessis AJ, Sullivan N, Khwaja OS, Zhang X, Barnes K, et al. Spectrum of neurodevelopmental disabilities in children with cerebellar malformations. Developmental Medicine and Child Neurology. 2011;53(5):409-416
  167. 167. Greiner MV, Lawrence AP, Horn P, Newmeyer AJ, Makoroff KL. Early clinical indicators of developmental outcome in abusive head trauma. Child's Nervous System. 2012;28(6):889-896
  168. 168. Marete I, Tenge C, Pasha O, Goudar S, Chomba E, Patel A, et al. Perinatal outcomes of multiple-gestation pregnancies in Kenya, Zambia, Pakistan, India, Guatemala, and Argentina: A global network study. American Journal of Perinatology. 2014;31(2):125-132
  169. 169. Nath S, Roy R, Mukherjee S. Perinatal complications associated with autism—A case control study in a neurodevelopment and early intervention clinic. Journal of the Indian Medical Association. 2012;110(8):526-529
  170. 170. François C, Ripollés P, Bosch L, Garcia-Alix A, Muchart J, Sierpowska J, et al. Language learning and brain reorganization in a 3.5-year-old child with left perinatal stroke revealed using structural and functional connectivity. Cortex. 2016;77:95-118
  171. 171. Young JM, Powell TL, Morgan BR, Card D, Lee W, Smith ML, et al. Deep grey matter growth predicts neurodevelopmental outcomes in very preterm children. NeuroImage. 2015;111:360-368. DOI: 10.1016/j.neuroimage.2015.02.030. Epub 2015 Feb 21
  172. 172. Roberts G, Cheong J, Opie G, et al. Growth of extremely preterm survivors from birth to 18 years of age compared with term controls. Pediatrics. 2013;131(2):e439-e445. DOI: 10.1542/peds.2012-1135.
  173. 173. Bocca-Tjeertes IFA, Kerstjens JM, Reijneveld SA, de Winter AF, Bos AF. Growth and predictors of growth restraint in moderately preterm children aged 0 to 4 years. Pediatrics. 2011;128(5):e1187-e11 94. DOI: 10.1542/peds.2010-3781
  174. 174. Bayer JK, Hiscock H, Ukoumunne OC, Price A, Wake M. Early childhood aetiology of mental health problems: A longitudinal population-based study. Journal of Child Psychology and Psychiatry. 2008;49(11):1166-1174
  175. 175. Huhtala M, Korja R, Lehtonen L, Haataja L, Lapinleimu H, Rautava P. Parental psychological well-being and behavioral outcome of very low birth weight infants at 3 years. Pediatrics. 2012;129(4):937-945
  176. 176. Embleton ND, Skeath T. Catch-up growth and metabolic and cognitive outcomes in adolescents born preterm. Nestle Nutrition Institute Workshop Series. 2015;81:61-71
  177. 177. Teller IC, Embleton ND, Griffin IJ, van Elburg RM. Post-discharge formula feeding in preterm infants: A systematic review mapping evidence about the role of macronutrient enrichment. Clinical Nutrition. 2015. pii: S0261-5614(15)00227-7. DOI: 10.1016/j.clnu.2015.08.006
  178. 178. Schmidt B, Roberts RS, Davis PG, et al. Prediction of late death or disability at age 5 years using a count of 3 neonatal morbidities in very low birth weight infants. The Journal of Pediatrics. 2015;167:982
  179. 179. Casolini P, Zuena AR, et al. Sub-neurotoxic neonatal anoxia induces subtle behavioural changes and specific abnormalities in brain group-I metabotropic glutamate receptors in rats. Journal of Neurochemistry. 2005;95:137-145
  180. 180. Padilla VT et al. Impact of severe intrauterine restriction on brain development. Acta Paediatrica. 2009;98(suppl. 460):204
  181. 181. Bjuland KJ, Rimol LM, Løhaugen GC, Skranes J. Brain volumes and cognitive function in very-low-birth-weight (VLBW) young adults. European Journal of Paediatric Neurology. 2014;18(5):578-590
  182. 182. Botellero VL, Skranes J, Bjuland KJ, Løhaugen GC, Håberg AK, Lydersen S, et al. Mental health and cerebellar volume during adolescence in very-low-birth-weight infants: A longitudinal study. Child and Adolescent Psychiatry and Mental Health. 2016;10:6. DOI: 10.1186/s13034-016-0093-8
  183. 183. Lees C, Marlow N, Arabin B, Bilardo CM, Brezinka C, Derks JB, et al. Perinatal morbidity and mortality in early-onset fetal growth restriction: Cohort outcomes of the trial of randomized umbilical and fetal flow in Europe (TRUFFLE). Ultrasound in Obstetrics & Gynecology. 2013;42:400-408
  184. 184. Pham H, Duy AP, Pansiot J, Bollen B, Gallego J, Charriaut-Marlangue C, et al. Impact of inhaled nitric oxide on white matter damage in growth-restricted neonatal rats. Pediatric Research. 2015;77(4):563-569
  185. 185. Hauglann L, Handegaard BH, Ulvund SE, Nordhov M, Rønning JA, Kaaresen PI. Cognitive outcome of early intervention in preterms at 7 and 9 years of age: A randomised controlled trial. Archives of Disease in Childhood. Fetal and Neonatal Edition. 2015;100(1):F11-F16
  186. 186. Van Wassenaer-Leemhuis AG, Jeukens-Visser M, van Hus JW, Meijssen D, Wolf MJ, Kok JH, et al. Rethinking preventive post-discharge intervention programmes for very preterm infants and their parents. Developmental Medicine and Child Neurology. 2016;58(Suppl 4):67-73
  187. 187. Swamy MK, Kamal P, Shailaja N. Maternal and perinatal outcome during expectant management. The Journal of Obstetrics and Gynecology of India. 2012;62(4):413-418

Written By

Jamila Gurbanova, Saadat Huseynova and Afat Hasanova

Submitted: 10 September 2022 Reviewed: 14 September 2022 Published: 02 November 2022