1
|
van de Sande MJA, Slaghekke F, Te Pas AB, Witlox RSGM, Lopriore E, Tollenaar LSA. Increased Risk of Persistent Pulmonary Hypertension of the Newborn in Twin Anaemia Polycythaemia Sequence Donors. Fetal Diagn Ther 2024:1-10. [PMID: 39532069 DOI: 10.1159/000542493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION This study aimed to describe the prevalence and risk factors for respiratory complications in monochorionic twins with twin anaemia polycythaemia sequence (TAPS). METHODS All neonates diagnosed with postnatal TAPS at our center between 2002 and 2023 were included in this retrospective study. The primary outcome was the prevalence of respiratory complications, including respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), and persistent pulmonary hypertension of the newborn (PPHN). Secondary outcomes included need of respiratory support during admission and a risk factor analysis for adverse respiratory outcome. RESULTS In our study of 100 postnatally diagnosed TAPS pregnancies, 32% (62/199) experienced RDS and 13% (25/199) had BPD, with no difference between donors and recipients. PPHN occurred in 7% of cases, more frequently in donors (11%, 11/100) than in recipients (3%, 3/100) (OR = 1.3, 95% CI: 0.2-2.6). Lower gestational age at birth and severe foetal anaemia were found to be significant independent risk factors associated with PPHN in TAPS twins (OR = 0.3, 95% CI: 0.1-0.5), respectively (OR = 1.9, 95% CI: 0.8-3.1). CONCLUSION TAPS donor twins have a fourfold increased risk of PPHN due to anaemia compared to recipient twins. Given the life-threatening nature of PPHN, TAPS twins should be born in hospitals equipped to treat it.
Collapse
Affiliation(s)
- Margot J A van de Sande
- Division of Fetal Therapy, Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Femke Slaghekke
- Division of Fetal Therapy, Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arjan B Te Pas
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ruben S G M Witlox
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Enrico Lopriore
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisanne S A Tollenaar
- Division of Fetal Therapy, Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Hu Y, Lin Y, Yang J, Wang S, Gao L, Bi Y, Wang Y. Mitochondrial dysfunction and oxidative stress in selective fetal growth restriction. Placenta 2024; 156:46-54. [PMID: 39265375 DOI: 10.1016/j.placenta.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
INTRODUCTION Placental dysfunction is the primary cause of selective fetal growth restriction (sFGR), and the specific role of mitochondria remains unclear. This study aims to elucidate mitochondrial functional defects in sFGR placentas and explore the roles of mitochondrial genomic and epigenetic alterations in its pathogenesis. METHODS The placental villi of MCDA twins with sFGR were collected and the morphology and number of mitochondria were observed by transmission electron microscopy. Meanwhile, the levels of reactive oxygen species (ROS), ATP and oxidative damage markers were assessed. Mitochondrial DNA (mtDNA) copy number detection, targeted sequencing and methylation sequencing were performed. The expression of placental cytochrome c oxidase subunit I (COX I) and mitochondrial long non-coding RNAs (lncRNAs) were evaluated by Western blotting and qPCR. RESULTS Compared with placentae from normal fetuses, pronounced mitochondrial damage within cytotrophoblast was revealed in sFGR placentae, alongside augmented mitochondrial number in syncytiotrophoblast. Enhanced oxidative stress in these placentae was evidenced by elevated markers of oxidative damage, accompanied by increased ROS production and diminished ATP generation. In sFGR placentae, a notable rise in mitochondrial copy number and one heterozygous mutation in the MT-RNR2 gene were observed, along with decreased COX Ⅰ levels, increased lncND5, lncND6, lncCyt b, and MDL1 synthesis, and decreased RMRP synthesis. DISCUSSION Findings collectively confirmed an exacerbation of oxidative stress within sFGR placentae, coinciding with mitochondrial dysfunction, compromised energy production, and ultimately the failure of compensatory mechanisms to restore energy balance, which may result from mutations in the mitochondrial genome and abnormal expression of epigenetic regulatory genes.
Collapse
Affiliation(s)
- Yucheng Hu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yuhong Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Jiawen Yang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Shan Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Li Gao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yan Bi
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| | - Yanlin Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| |
Collapse
|
3
|
El Emrani S, Jansen EJS, Goeman JJ, Termote JUM, Lopriore E, Schalij-Delfos NE, van der Meeren LE. Enhancing the Retinopathy Of Prematurity Risk Profile Through Placental Evaluation of Maternal and Fetal Vascular Malperfusion. Invest Ophthalmol Vis Sci 2024; 65:9. [PMID: 39230991 PMCID: PMC11379085 DOI: 10.1167/iovs.65.11.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Purpose To determine the independent effect of uteroplacental malperfusion on the development of retinopathy of prematurity (ROP). Methods This cohort study included 591 neonates with a gestational age (GA) ≤ 32 weeks or birthweight (BW) ≤ 1500 g. Clinical data was retrospectively collected and placentas were prospectively examined for maternal vascular malperfusion (e.g., abruption, infarct, distal villous hypoplasia, ischemia, and decidual necrosis) and fetal vascular malperfusion (e.g., thrombosis, fetal hypoxia, and hydrops parenchyma). The primary outcome was ROP. Secondary outcomes were GA, BW, small for gestational age (SGA), mechanical ventilation duration, postnatal corticosteroids, sepsis, and necrotizing enterocolitis. Results Maternal vascular malperfusion was associated with higher GA, lower BW, and increased SGA rates, except placental abruption, which was associated with lower SGA rates. Fetal vascular malperfusion was associated with lower BW, increased SGA rates and lower duration of mechanical ventilation. Subgroup analysis of placentas without inflammation showed increased rates of distal villous hypoplasia (44% vs. 31%) and hydrops parenchyma (7% vs. 0%) in neonates with ROP. Multivariate regression analyses revealed three placenta factors to be independently associated with ROP: distal villous hypoplasia (OR = 1.7; 95% CI, 1.0-3.0), severe acute histological chorioamnionitis (OR = 2.1; 95% CI, 1.1-3.9) and funisitis (OR = 1.8; 95% CI, 1.0-3.1). Conclusions Placental evaluation of distal villous hypoplasia, severe acute chorioamnionitis and funisitis is a novel and valuable addition to the ROP risk profile. Evaluation of these placental risk factors shortly after birth can aid in identifying high-risk infants in an earlier stage than currently possible.
Collapse
Affiliation(s)
- Salma El Emrani
- Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther J S Jansen
- Neonatology, Wilhelmina Children's Hospital, Department of Women and Neonate, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jelle J Goeman
- Medical statistics, Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacqueline U M Termote
- Neonatology, Wilhelmina Children's Hospital, Department of Women and Neonate, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Enrico Lopriore
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Lotte E van der Meeren
- Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Fuentes Carballal JA, Pumarada Prieto M, Crespo Suárez PA, Luaces González J, López Conde I, Picans Leis R, Sardina Ríos A, Durán Fernández-Feijoo C, Avila-Alvarez A. Impact on neonatal morbidity of moderate to severe early foetal growth restriction defined by doppler criteria: multicentre study. An Pediatr (Barc) 2024; 101:104-114. [PMID: 39138032 DOI: 10.1016/j.anpede.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/19/2024] [Indexed: 08/15/2024] Open
Abstract
INTRODUCTION In recent years, there has been a change in the conceptualization of foetal growth restriction (FGR), which has gone from being defined solely based on weight criteria to being defined and staged based on Doppler criteria. The aim of our study was to evaluate neonatal risk in a cohort of neonates with moderate to severe early-onset FGR defined by Doppler criteria. POPULATION AND METHODS We conducted a multicentre prospective cohort study in a cohort of neonates with early-onset foetal growth restriction and abnormal Doppler findings and a control cohort without Doppler abnormalities matched for sex and gestational age. RESULTS A total of 105 patients (50 cases, 55 controls) were included. We found a higher frequency of respiratory morbidity in the FGR group, with an increased need of surfactant (30% vs. 27.3%; OR, 5.3 [95% CI, 1.1-26.7]), an increased need for supplemental oxygen (66% vs. 49.1%; OR, 5.6 [95% CI, 1.5-20.5]), and a decreased survival without bronchopulmonary dysplasia (70 vs. 87.3%; OR, 0.16 [95% CI, 0.03-0.99]). Patients with FGR required a longer length of stay and more days of parenteral nutrition and had a higher incidence of haematological abnormalities such as neutropenia and thrombopenia. The lactate level at birth was higher in the severe FGR subgroup (6.12 vs. 2.4 mg/dL; P = .02). CONCLUSION The diagnosis of early-onset moderate to severe FGR defined by Doppler criteria carries a greater risk of respiratory, nutritional and haematological morbidity, independently of weight and gestational age. These patients, therefore, should be considered at increased risk compared to constitutionally small for gestational age preterm infants or preterm infants without FGR.
Collapse
Affiliation(s)
- Jesús Alberto Fuentes Carballal
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Universitario de A Coruña, Servizo Galego de Saúde (SERGAS), A Coruña, Spain.
| | - Marcelino Pumarada Prieto
- Unidad de Neonatología, Servicio de Pediatría, Hospital Álvaro Cunqueiro, Servizo Galego de Saúde (SERGAS), Vigo, Spain
| | - Pilar Adelaida Crespo Suárez
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Pontevedra (CHOP), Servizo Galego de Saúde (SERGAS), Pontevedra, Spain
| | - José Luaces González
- Servicio de Pediatría, Hospital Arquitecto Marcide, Servizo Galego de Saúde (SERGAS), Ferrol, Spain
| | - Isabel López Conde
- Unidad de Neonatología, Servicio de Pediatría, Hospital Lucus Augusti, Servizo Galego de Saúde (SERGAS), Lugo, Spain
| | - Rosaura Picans Leis
- Servicio de Pediatría, Complexo Hospitalario Universitario de Santiago (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Alicia Sardina Ríos
- Unidad Neonatología, Servicio de Pediatría, Complexo Hospitalario Ourense (CHOU), Servizo Galego de Saúde (SERGAS), Ourense, Spain
| | | | - Alejandro Avila-Alvarez
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Universitario de A Coruña, Servizo Galego de Saúde (SERGAS), A Coruña, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| |
Collapse
|
5
|
Wu TJ, Jing X, Teng M, Pritchard KA, Day BW, Naylor S, Teng RJ. Role of Myeloperoxidase, Oxidative Stress, and Inflammation in Bronchopulmonary Dysplasia. Antioxidants (Basel) 2024; 13:889. [PMID: 39199135 PMCID: PMC11351552 DOI: 10.3390/antiox13080889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 09/01/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a lung complication of premature births. The leading causes of BPD are oxidative stress (OS) from oxygen treatment, infection or inflammation, and mechanical ventilation. OS activates alveolar myeloid cells with subsequent myeloperoxidase (MPO)-mediated OS. Premature human neonates lack sufficient antioxidative capacity and are susceptible to OS. Unopposed OS elicits inflammation, endoplasmic reticulum (ER) stress, and cellular senescence, culminating in a BPD phenotype. Poor nutrition, patent ductus arteriosus, and infection further aggravate OS. BPD survivors frequently suffer from reactive airway disease, neurodevelopmental deficits, and inadequate exercise performance and are prone to developing early-onset chronic obstructive pulmonary disease. Rats and mice are commonly used to study BPD, as they are born at the saccular stage, comparable to human neonates at 22-36 weeks of gestation. The alveolar stage in rats and mice starts at the postnatal age of 5 days. Because of their well-established antioxidative capacities, a higher oxygen concentration (hyperoxia, HOX) is required to elicit OS lung damage in rats and mice. Neutrophil infiltration and ER stress occur shortly after HOX, while cellular senescence is seen later. Studies have shown that MPO plays a critical role in the process. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), a reversible MPO inhibitor, attenuates BPD effectively. In contrast, the irreversible MPO inhibitor-AZD4831-failed to provide similar efficacy. Interestingly, KYC cannot offer its effectiveness without the existence of MPO. We review the mechanisms by which this anti-MPO agent attenuates BPD.
Collapse
Affiliation(s)
- Tzong-Jin Wu
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Xigang Jing
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Michelle Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Kirkwood A. Pritchard
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
- Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Billy W. Day
- ReNeuroGen LLC, 2160 San Fernando Dr, Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Stephen Naylor
- ReNeuroGen LLC, 2160 San Fernando Dr, Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Ru-Jeng Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| |
Collapse
|
6
|
Rebai N, Lopriore E, Bekker V, Slaghekke F, Schoenaker MHD, Groene SG. Necrotizing enterocolitis in monochorionic twins: Insights from an identical twin model. Early Hum Dev 2024; 194:106052. [PMID: 38781714 DOI: 10.1016/j.earlhumdev.2024.106052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Necrotizing enterocolitis (NEC) is a major cause of neonatal morbidity and mortality in preterm neonates, yet its pathophysiology remains unclear. The aim of this study is to evaluate risk factors for NEC using an identical twin model. In this case-control study, all monochorionic twin pairs born in our center in 2002-2020 were retrospectively reviewed for NEC. Potential risk factors for NEC were studied. For within-pair comparison, outcomes were compared between affected and unaffected twins. Within-pair analyses showed that the twin with NEC had a lower birth weight compared to its unaffected co-twin (1100 (913-1364) vs. 1339 (1093-1755) grams). Median gestational age at birth and birth weight were lower in twin pairs in the NEC-group compared to the no-NEC group, 29.1 weeks (27.8-30.8) versus 33.6 (30.7-36.0) and 1221 g (1010-1488) versus 1865 (1356-2355) respectively. Twin pregnancies in the NEC-group were more often complicated by twin-to-twin transfusion syndrome compared to the no-NEC-group (70 % (14/20) vs. 49 % (472/962)), particularly when treated with amnioreduction. This unique population of identical twins confirms that preterm neonates with a relatively lower birth weight are more prone to develop NEC compared to their co-twin, regardless of other genetic, maternal and obstetrical factors.
Collapse
Affiliation(s)
- Nour Rebai
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Enrico Lopriore
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Vincent Bekker
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Femke Slaghekke
- Department of Obstetrics, Division of Fetal Therapy, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Michiel H D Schoenaker
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Willem-Alexander Children's Hospital, Laboratory for Pediatric Immunology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Sophie G Groene
- Neonatoloy, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
7
|
樊 雨, 张 伊, 温 和, 晏 红, 沈 蔚, 丁 月, 龙 运, 张 志, 李 桂, 姜 泓, 饶 红, 邱 建, 魏 贤, 张 亚, 曾 纪, 赵 常, 许 伟, 王 凡, 员 丽, 杨 秀, 李 薇, 林 霓, 陈 倩, 夏 昌, 钟 鑫, 崔 其. [Risk factors for bronchopulmonary dysplasia in twin preterm infants: a multicenter study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:611-618. [PMID: 38926378 PMCID: PMC11562066 DOI: 10.7499/j.issn.1008-8830.2312005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/08/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVES To investigate the risk factors for bronchopulmonary dysplasia (BPD) in twin preterm infants with a gestational age of <34 weeks, and to provide a basis for early identification of BPD in twin preterm infants in clinical practice. METHODS A retrospective analysis was performed for the twin preterm infants with a gestational age of <34 weeks who were admitted to 22 hospitals nationwide from January 2018 to December 2020. According to their conditions, they were divided into group A (both twins had BPD), group B (only one twin had BPD), and group C (neither twin had BPD). The risk factors for BPD in twin preterm infants were analyzed. Further analysis was conducted on group B to investigate the postnatal risk factors for BPD within twins. RESULTS A total of 904 pairs of twins with a gestational age of <34 weeks were included in this study. The multivariate logistic regression analysis showed that compared with group C, birth weight discordance of >25% between the twins was an independent risk factor for BPD in one of the twins (OR=3.370, 95%CI: 1.500-7.568, P<0.05), and high gestational age at birth was a protective factor against BPD (P<0.05). The conditional logistic regression analysis of group B showed that small-for-gestational-age (SGA) birth was an independent risk factor for BPD in individual twins (OR=5.017, 95%CI: 1.040-24.190, P<0.05). CONCLUSIONS The development of BPD in twin preterm infants is associated with gestational age, birth weight discordance between the twins, and SGA birth.
Collapse
Affiliation(s)
| | | | - 和梅 温
- 四川锦欣妇女儿童医院新生儿科,四川成都610011
| | - 红 晏
- 贵州医科大学附属医院新生儿科,贵州贵阳550001
| | - 蔚 沈
- 厦门大学附属妇女儿童医院新生儿科,福建厦门361003
| | - 月琴 丁
- 南方医科大学附属东莞医院新生儿科,广东东莞523000
| | - 运峰 龙
- 邵阳学院附属第一医院新生儿科,湖南邵阳422000
| | | | | | | | | | - 建武 邱
- 汕头大学医学院附属粤北人民医院新生儿科,广东韶关512026
| | - 贤 魏
- 武汉科技大学附属孝感医院新生儿科,湖北孝感432000
| | - 亚昱 张
- 内蒙古医科大学附属医院新生儿科,内蒙古呼和浩特010050
| | - 纪斌 曾
- 汕头大学医学院第二附属医院新生儿科,广东汕头515041
| | - 常亮 赵
- 包钢集团第三;职工医院新生儿科,内蒙古包头014010
| | - 伟鹏 许
- 暨南大学附属第一医院新生儿科,广东广州510630
| | | | | | | | - 薇 李
- 东莞市滨海湾中心医院新生儿科,广东东莞523808
| | - 霓阳 林
- 汕头大学;医学院第一附属医院新生儿科,广东汕头515041
| | | | | | | | | |
Collapse
|
8
|
Spekman JA, Israëls J, de Vreede I, Los M, Geelhoed MJ, van Zwet EW, Haak MC, Roest AA, van Klink JM, Lopriore E, Groene SG. Reduced lung function during childhood in identical twins with discordant fetal growth: a cohort study. EClinicalMedicine 2024; 72:102600. [PMID: 38633574 PMCID: PMC11019090 DOI: 10.1016/j.eclinm.2024.102600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
Background Fetal growth restriction (FGR) can negatively affect lung development, leading to increased respiratory morbidity and reduced lung function later in life. Studies regarding the impact of FGR on lung function in singletons are influenced by genetic, obstetric, and maternal factors. To overcome these confounding factors, we aim to investigate lung function in identical twins with selective FGR (sFGR). Methods Lung function assessments were performed in identical twins with sFGR born in our centre between March 1, 2002, and December 31, 2017, aged between 5 and 17 years. sFGR was defined as birthweight discordance ≥20%. Outcome measures consisted of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC), and transfer factor for carbon monoxide (DLCO) and were compared between the smaller and larger twin. Findings Thirty-nine twin pairs performed spirometry of sufficient quality. Median gestational age at birth was 34.3 (interquartile range (IQR) 32.1-36.0) weeks with median birthweights of 1500 (IQR 1160-1880) grams and 2178 (IQR 1675-2720) grams for the smaller and larger twin, respectively. Smaller twins had significantly lower z-scores for FEV1 (-0.94 versus -0.41, p = 0.0015), FVC (-0.56 versus -0.06, p < 0.0001) and DLCO (-0.50 versus 0.00, p < 0.0001) compared to larger co-twins. Interpretation Although being genetically identical, sFGR in identical twins is associated with a reduction in static and dynamic lung volume and a reduction in lung diffusion, even when taking the reduced lung volume into account. This indicates that adverse growth conditions in utero negatively affect lung development and function, potentially contributing to an increase in respiratory morbidities later in life. Funding The Dutch Heart Foundation and The Bontius Foundation.
Collapse
Affiliation(s)
- Jip A. Spekman
- Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, the Netherlands
| | - Joël Israëls
- Pediatric Pulmonology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ilja de Vreede
- Pediatric Pulmonology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mady Los
- Department of Pulmonology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Miranda J.J. Geelhoed
- Department of Pulmonology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Erik W. van Zwet
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Monique C. Haak
- Fetal Therapy, Department of Obstetrics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Arno A.W. Roest
- Pediatric Cardiology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jeanine M.M. van Klink
- Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, the Netherlands
| | - Enrico Lopriore
- Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, the Netherlands
| | - Sophie G. Groene
- Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
9
|
Gounaris AK, Sokou R. Nutrition and Growth of Preterm Neonates during Hospitalization: Impact on Childhood Outcomes. Nutrients 2024; 16:218. [PMID: 38257111 PMCID: PMC10819760 DOI: 10.3390/nu16020218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
The Special Issue has been completed with the publication of 13 review and research articles [...].
Collapse
Affiliation(s)
- Antonios K. Gounaris
- Neonatal Intensive Care Unit, University Hospital of Larissa, 413 34 Larissa, Greece;
| | - Rozeta Sokou
- Neonatal Intensive Care Unit, Nikea General Hospital “Agios Panteleimon”, 184 54 Piraeus, Greece
| |
Collapse
|
10
|
Anciuc-Crauciuc M, Cucerea MC, Tripon F, Crauciuc GA, Bănescu CV. Descriptive and Functional Genomics in Neonatal Respiratory Distress Syndrome: From Lung Development to Targeted Therapies. Int J Mol Sci 2024; 25:649. [PMID: 38203821 PMCID: PMC10780183 DOI: 10.3390/ijms25010649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
In this up-to-date study, we first aimed to highlight the genetic and non-genetic factors associated with respiratory distress syndrome (RDS) while also focusing on the genomic aspect of this condition. Secondly, we discuss the treatment options and the progressing therapies based on RNAs or gene therapy. To fulfill this, our study commences with lung organogenesis, a highly orchestrated procedure guided by an intricate network of conserved signaling pathways that ultimately oversee the processes of patterning, growth, and differentiation. Then, our review focuses on the molecular mechanisms contributing to both normal and abnormal lung growth and development and underscores the connections between genetic and non-genetic factors linked to neonatal RDS, with a particular emphasis on the genomic aspects of this condition and their implications for treatment choices and the advancing therapeutic approaches centered around RNAs or gene therapy.
Collapse
Affiliation(s)
- Mădălina Anciuc-Crauciuc
- Genetics Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540142 Târgu Mureș, Romania; (M.A.-C.); (C.V.B.)
- Neonatology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540142 Târgu Mureș, Romania;
| | - Manuela Camelia Cucerea
- Neonatology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540142 Târgu Mureș, Romania;
| | - Florin Tripon
- Genetics Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540142 Târgu Mureș, Romania; (M.A.-C.); (C.V.B.)
| | - George-Andrei Crauciuc
- Genetics Laboratory, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania;
| | - Claudia Violeta Bănescu
- Genetics Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540142 Târgu Mureș, Romania; (M.A.-C.); (C.V.B.)
- Genetics Laboratory, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania;
| |
Collapse
|
11
|
Wu ZH, Li FF, Ruan LL, Feng Q, Zhang S, Li ZH, Otoo A, Tang J, Fu LJ, Liu TH, Ding YB. miR-181d-5p, which is upregulated in fetal growth restriction placentas, inhibits trophoblast fusion via CREBRF. J Assist Reprod Genet 2023; 40:2725-2737. [PMID: 37610607 PMCID: PMC10643557 DOI: 10.1007/s10815-023-02917-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/15/2023] [Indexed: 08/24/2023] Open
Abstract
PURPOSE Fetal growth restriction (FGR) is a common complication characterized by impaired placental function and unfavorable pregnancy outcomes. This study aims to elucidate the expression pattern of miR-181d-5p in FGR placentas and explore its effects on trophoblast fusion. METHODS The expression pattern of miR-181d-5p in human FGR placentas were evaluated using qRT-PCR. Western blot, qRT-PCR, and Immunofluorescence analysis were performed in a Forskolin (FSK)-induced BeWo cell fusion model following the transfection of miR-181d-5p mimic or inhibitor. Potential target genes for miR-181d-5p were identified by screening miRNA databases. The interaction between miR-181d-5p and Luman/CREB3 Recruitment Factor (CREBRF) was determined through a luciferase assay. Moreover, the effect of CREBRF on BeWo cell fusion was examined under hypoxic conditions. RESULTS Aberrant up-regulation of miR-181d-5p and altered expression of trophoblast fusion makers, including glial cell missing 1 (GCM1), Syncytin1 (Syn1), and E-cadherin (ECAD), were found in human FGR placentas. A down-regulation of miR-181d-5p expression was observed in the FSK-induced BeWo cell fusion model. Transfection of the miR-181d-5p mimic resulted in the inhibition of BeWo cell fusion, characterized by a down-regulation of GCM1 and Syn1, accompanied by an up-regulation of ECAD. Conversely, the miR-181d-5p inhibitor promoted BeWo cell fusion. Furthermore, miR-181d-5p exhibited negative regulation of CREBRF, which was significantly down-regulated in the hypoxia-induced BeWo cell model. The overexpression of CREBRF was effectively ameliorated the impaired BeWo cell fusion induced by hypoxia. CONCLUSIONS Our study demonstrated that miR-181d-5p, which is elevated in FGR placenta, inhibited the BeWo cell fusion through negatively regulating the expression of CREBRF.
Collapse
Affiliation(s)
- Zhi-Hong Wu
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, No.120 Longshan Road, Yubei District, Chongqing, 401147, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China
| | - Fang-Fang Li
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China
| | - Ling-Ling Ruan
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China
| | - Qian Feng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Shuang Zhang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China
| | - Zhuo-Hang Li
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China
| | - Antonia Otoo
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China
| | - Jing Tang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China
| | - Li-Juan Fu
- Department of Pharmacology, the School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China.
- Academician Workstation, Changsha Medical University, Changsha, China.
| | - Tai-Hang Liu
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China.
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, No.120 Longshan Road, Yubei District, Chongqing, 401147, China.
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Spekman JA, El Emrani S, Schalij-Delfos NE, Slaghekke F, van Klink JMM, Lopriore E, Groene SG. Association between fetal growth-restriction and retinopathy of prematurity using a unique identical twin model. Pediatr Res 2023; 94:1738-1743. [PMID: 37258717 DOI: 10.1038/s41390-023-02670-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/24/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Research in singletons identified fetal growth restriction (FGR) as a risk factor for retinopathy of prematurity (ROP), but is generally subject to confounding by genetic, obstetric, and maternal factors. We investigated the effect of FGR on ROP in growth-discordant identical twins, thereby controlling for confounding factors. METHODS All data of monochorionic (MC) twin pairs with a birth weight discordance ≥20% born in our center between 2010 and 2021 were retrospectively reviewed for the presence of ROP. Potential risk factors for ROP were analyzed. Outcomes were compared between the smaller and larger twin. RESULTS We included 88 MC twin pairs with growth discordance. In 34% (30/88), both neonates were at risk of ROP. Prevalence of ROP was higher among the smaller twin compared to the larger twin, 30% (9/30) versus 13% (4/30), respectively (OR 2.8, 95% CI: 1.2-6.6). The smaller twin had a longer duration of mechanical ventilation (8 (1-20) versus 2 (1-4) days) and received their first red blood cell transfusion at an earlier postmenstrual age (29.6 (28.1-31.6) versus 30.4 (29.7-32.6) weeks). CONCLUSIONS In this identical twin model, FGR is associated with almost tripled odds of ROP development, suggesting that both unfavorable antenatal growth conditions and adverse neonatal outcomes affect postnatal retinal vascular proliferation. IMPACT Fetal growth restriction in growth-discordant identical twins is associated with almost tripled odds of developing retinopathy of prematurity in the smaller twin. Since these twins do not only differ in birth weight but also duration of mechanical ventilation and timing of the first red blood cell transfusion, both unfavorable antenatal growth conditions and adverse neonatal outcomes can affect postnatal retinal vascular proliferation. More attention for preventing retinopathy of prematurity is needed in those with fetal growth restriction who received prolonged duration of mechanical ventilation, oxygen supplementation, or a first red blood cell transfusion <32 weeks postmenstrual age.
Collapse
Affiliation(s)
- Jip A Spekman
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Salma El Emrani
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Femke Slaghekke
- Fetal Therapy, Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeanine M M van Klink
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Enrico Lopriore
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sophie G Groene
- Neonatology, Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Gounaris AK, Sokou R, Gounari EA, Panagiotounakou P, Grivea IN. Extrauterine Growth Restriction and Optimal Growth of Very Preterm Neonates: State of the Art. Nutrients 2023; 15:3231. [PMID: 37513649 PMCID: PMC10384682 DOI: 10.3390/nu15143231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Over the last few decades, there has been an ongoing debate over both the optimal feeding mode for very premature neonates (VPN) as well as what their optimal growth should be. Despite the American Academy of Pediatric declaring since 1997 that the growth of VPN should follow the trajectory of intrauterine fetal growth, differences of opinion persist, feeding policies keep changing, and the growth and development of VPN remains extremely variable not only between countries, but even between neighboring neonatal units. Even the appropriate terminology to express poor postnatal growth (extrauterine growth restriction (EGR) and postnatal growth failure (PGF)) remains a subject of ongoing discussion. A number of recent publications have shown that by implementing breast milk fortification and closely following growth and adjusting nutrition accordingly, as per the consensus guidelines of the major Neonatal Societies, we could achieve growth that closely follows birth centiles. A recent position paper from EPSGAN recommending targeted nutritional support to cover the energy and protein deficits sustained by VPN during periods of critical illness further strengthens the above findings. Conclusion: We can promote better growth of VPN by ensuring a stable administration of sufficient calories and protein, especially in the first 2 weeks of life, implementing breast milk fortification, covering energy and protein deficits due to critical illness, and increasing feeding volumes as per the latest guidelines. The adoption of universal protocol for nutrition and growth of VPN is essential and will enable better monitoring of long-term outcomes for this population.
Collapse
Affiliation(s)
- Antonios K Gounaris
- Neonatal Clinic-NICU, University Hospital of Larissa, 413 34 Larissa, Greece
| | - Rozeta Sokou
- Neonatal Clinic-NICU, Nikea General Hospital "Agios Panteleimon", 184 54 Piraeus, Greece
| | | | | | - Ioanna N Grivea
- Neonatal Clinic-NICU, University Hospital of Larissa, 413 34 Larissa, Greece
| |
Collapse
|
14
|
Jang W, Choi YS, Kim JY, Yon DK, Lee YJ, Chung SH, Kim CY, Yeo SG, Lee J. Artificial Intelligence-Driven Respiratory Distress Syndrome Prediction for Very Low Birth Weight Infants: Korean Multicenter Prospective Cohort Study. J Med Internet Res 2023; 25:e47612. [PMID: 37428525 PMCID: PMC10366668 DOI: 10.2196/47612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/04/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND Respiratory distress syndrome (RDS) is a disease that commonly affects premature infants whose lungs are not fully developed. RDS results from a lack of surfactant in the lungs. The more premature the infant is, the greater is the likelihood of having RDS. However, even though not all premature infants have RDS, preemptive treatment with artificial pulmonary surfactant is administered in most cases. OBJECTIVE We aimed to develop an artificial intelligence model to predict RDS in premature infants to avoid unnecessary treatment. METHODS In this study, 13,087 very low birth weight infants who were newborns weighing less than 1500 grams were assessed in 76 hospitals of the Korean Neonatal Network. To predict RDS in very low birth weight infants, we used basic infant information, maternity history, pregnancy/birth process, family history, resuscitation procedure, and test results at birth such as blood gas analysis and Apgar score. The prediction performances of 7 different machine learning models were compared, and a 5-layer deep neural network was proposed in order to enhance the prediction performance from the selected features. An ensemble approach combining multiple models from the 5-fold cross-validation was subsequently developed. RESULTS Our proposed ensemble 5-layer deep neural network consisting of the top 20 features provided high sensitivity (83.03%), specificity (87.50%), accuracy (84.07%), balanced accuracy (85.26%), and area under the curve (0.9187). Based on the model that we developed, a public web application that enables easy access for the prediction of RDS in premature infants was deployed. CONCLUSIONS Our artificial intelligence model may be useful for preparations for neonatal resuscitation, particularly in cases involving the delivery of very low birth weight infants, as it can aid in predicting the likelihood of RDS and inform decisions regarding the administration of surfactant.
Collapse
Affiliation(s)
- Woocheol Jang
- Biomedical Engineering, Kyung Hee University, Yongin-si, Republic of Korea
| | - Yong Sung Choi
- Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Ji Yoo Kim
- Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Dong Keon Yon
- Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Young Joo Lee
- Department of Obstetrics and Gynecology, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Sung-Hoon Chung
- Department of Pediatrics, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Chae Young Kim
- Department of Pediatrics, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology Head and Neck Surgery, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Jinseok Lee
- Biomedical Engineering, Kyung Hee University, Yongin-si, Republic of Korea
| |
Collapse
|
15
|
Dankhara N, Holla I, Ramarao S, Kalikkot Thekkeveedu R. Bronchopulmonary Dysplasia: Pathogenesis and Pathophysiology. J Clin Med 2023; 12:4207. [PMID: 37445242 DOI: 10.3390/jcm12134207] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD), also known as chronic lung disease, is the most common respiratory morbidity in preterm infants. "Old" or "classic" BPD, as per the original description, is less common now. "New BPD", which presents with distinct clinical and pathological features, is more frequently observed in the current era of advanced neonatal care, where extremely premature infants are surviving because of medical advancements. The pathogenesis of BPD is complex and multifactorial and involves both genetic and environmental factors. This review provides an overview of the pathology of BPD and discusses the influence of several prenatal and postnatal factors on its pathogenesis, such as maternal factors, genetic susceptibility, ventilator-associated lung injury, oxygen toxicity, sepsis, patent ductus arteriosus (PDA), and nutritional deficiencies. This in-depth review draws on existing literature to explore these factors and their potential contribution to the development of BPD.
Collapse
Affiliation(s)
- Nilesh Dankhara
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Ira Holla
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Sumana Ramarao
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | |
Collapse
|
16
|
Zhuxiao R, Fang X, Wei W, Shumei Y, Jianlan W, Qiuping L, Jingjun P, Chuan N, Yongsheng L, Zhichun F, Jie Y. Prevention for moderate or severe BPD with intravenous infusion of autologous cord blood mononuclear cells in very preterm infants-a prospective non-randomized placebo-controlled trial and two-year follow up outcomes. EClinicalMedicine 2023; 57:101844. [PMID: 36864985 PMCID: PMC9971511 DOI: 10.1016/j.eclinm.2023.101844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is the primary severe complication of preterm birth. Severe BPD was associated with higher risks of mortality, more postnatal growth failure, long term respiratory and neurological developmental retardation. Inflammation plays a central role in alveolar simplification and dysregulated vascularization of BPD. There is no effective treatment to improve BPD severity in clinical practice. Our previous clinical study showed autologous cord blood mononuclear cells (ACBMNCs) infusion could reduce the respiratory support duration safely and potential improved BPD severity. Abundant preclinical studies have reported the immunomodulation effect as an important mechanism underlying the beneficial results of stem cell therapies in preventing and treating BPD. However, clinical studies assessing the immunomodulatory effect after stem cells therapy were rare. This study was to investigate the effect of ACBMNCs infusion soon after birth on prevention for severe BPD and long term outcomes in very preterm neonates. The immune cells and inflammatory biomarkers were detected to investigate the underlying immunomodulatory mechanisms. METHODS This single-center, prospective, investigator-initiated, non-randomized trial with blinded outcome assessment aimed to assess the effect of a single intravenous infusion of ACBMNCs in preventing severe BPD (moderate or severe BPD at 36 weeks of gestational age or discharge home) in surviving very preterm neonates less than 32 gestational weeks. Patients admitted to the Neonatal Intensive Care Unit (NICU) of Guangdong Women and Children Hospital from July 01, 2018 to January 1, 2020 were assigned to receiving a targeted dosage of 5 × 107 cells/kg ACBMNC or normal saline intravenously within 24 h after enrollment. Incidence of moderate or severe BPD in survivors were investigated as the primary short term outcome. Growth, respiratory and neurological development were assessed as long term outcomes at corrected age of 18-24 month-old. Immune cells and inflammatory biomarkers were detected for potential mechanism investigation. The trial was registered at ClinicalTrials.gov (NCT02999373). FINDINGS Six-two infants were enrolled, of which 29 were enrolled to intervention group, 33 to control group. Moderate or severe BPD in survivors significantly decreased in intervention group (adjusted p = 0.021). The number of patients needed to treat to gain one moderate or severe BPD-free survival was 5 (95% confidence interval: 3-20). Survivors in the intervention group had a significantly higher chance to be extubated than infants in the control group (adjusted p = 0.018). There was no statistical significant difference in total BPD incidence (adjusted p = 0.106) or mortality (p = 1.000). Incidence of developmental delay reduced in intervention group in long term follow-up (adjusted p = 0.047). Specific immune cells including proportion of T cells (p = 0.04) and CD4+ T cells in lymphocytes (p = 0.03), and CD4+ CD25+ forkhead box protein 3 (FoxP3)+ regulatory T cells in CD4+ T cells increased significantly after ACBMNCs intervention (p < 0.001). Anti-inflammatory factor IL-10 was higher (p = 0.03), while pro-inflammatory factor such as TNF-a (p = 0.03) and C reactive protein (p < 0.001) level was lower in intervention group than in control group after intervention. INTERPRETATION ACBMNCs could prevent moderate or severe BPD in surviving very premature neonates and might improve neurodevelopmental outcomes in long term. An immunomodulatory effect of MNCs contributed to the improvement of BPD severity. FUNDING This work was supported by National Key R&D Program of China (2021YFC2701700), National Natural Science Foundation of China (82101817, 82171714, 8187060625), Guangzhou science and technology program (202102080104).
Collapse
Affiliation(s)
- Ren Zhuxiao
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xu Fang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wei Wei
- Guang Dong Cord Blood Bank, Guangzhou, China
| | - Yang Shumei
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wang Jianlan
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Li Qiuping
- Department of Neonatology, The 7th Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
| | - Pei Jingjun
- Department of Neonatology, Nanfang Hospital, Southern Medical University, China
| | - Nie Chuan
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | | | - Feng Zhichun
- Department of Neonatology, The 7th Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Corresponding author. Department of Neonatology, The 7th Medical Center of PLA General Hospital, Beijing, China.
| | - Yang Jie
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
- Department of Neonatology, Nanfang Hospital, Southern Medical University, China
- Corresponding author. Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China; Department of Neonatology, Nanfang Hospital, Southern Medical University, China.
| |
Collapse
|
17
|
Groene SG, Jansen L, Tan RNGB, Steggerda SJ, Haak MC, Roest AAW, Lopriore E, van Klink JMM. Insecure attachment and internalizing behavior problems in growth discordant identical twins. Early Hum Dev 2022; 174:105679. [PMID: 36179588 DOI: 10.1016/j.earlhumdev.2022.105679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Psychosocial development in monochorionic (MC) twins born after selective fetal growth restriction (sFGR) has been unreported to date, despite its importance for daily functioning and future relationships. AIMS To investigate psychosocial development, attachment and school functioning in MC twins with sFGR and compare outcomes with the general population and between smaller and larger twins. STUDY DESIGN Observational cohort study. SUBJECTS MC twins with sFGR (defined as a birth weight discordance ≥20 %) born between 2002 and 2017 and aged 3-17 years. OUTCOME MEASURES Multiple parent report questionnaires: the Child Behavior Checklist (social-emotional development and behavior), the (Early) Childhood Behavior Questionnaire Very Short Form (temperament), the Attachment Insecurity Screening Inventory (attachment) and a school functioning questionnaire. RESULTS Median age for the 48 twin pairs was 11 (interquartile range (IQR) 8-13) years. Attachment insecurity for both twins was higher than in the general population for ambivalence/resistance (34 % (21/62) vs. 16 %, p = 0.024) and total attachment insecurity (35 % (22/62) vs. 16 %, p = 0.016). Smaller twins had more internalizing behavioral problems, i.e. negative emotions and behaviors turned inwards (22 % (10/46) vs. 11 % (5/46), p = 0.021) and a higher negative affect, i.e. more likely to experience negative emotions (3.2 (2.9-3.7) vs. 2.9 (2.2-3.2), p = 0.009) than larger twins, as well as a lower secondary school level (p = 0.031). CONCLUSION MC twins with sFGR have more ambivalent/resistant attachment insecurity following the complicated pregnancy course. Smaller twins have a tendency towards negative emotions and internalizing behaviors compared to larger twins, indicating an increased sensitivity for depression and anxiety.
Collapse
Affiliation(s)
- Sophie G Groene
- Neonatology, Willem-Alexander Children's Hospital, Dept. of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands; Molecular Epidemiology, Dept. of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands.
| | - Lisette Jansen
- Dept. of Medical Psychology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ratna N G B Tan
- Neonatology, Willem-Alexander Children's Hospital, Dept. of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sylke J Steggerda
- Neonatology, Willem-Alexander Children's Hospital, Dept. of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Monique C Haak
- Fetal Therapy, Dept. of Obstetrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Arno A W Roest
- Pediatric Cardiology, Willem-Alexander Children's Hospital, Dept. of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Enrico Lopriore
- Neonatology, Willem-Alexander Children's Hospital, Dept. of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeanine M M van Klink
- Neonatology, Willem-Alexander Children's Hospital, Dept. of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
18
|
Groene SG, Openshaw KM, Jansén-Storbacka LR, Slaghekke F, Haak MC, Heijmans BT, van Klink JMM, Roest AAW, van der Meeren LE, Lopriore E. Impact of placental sharing and large bidirectional anastomoses on birthweight discordance in monochorionic twins: a retrospective cohort study in 449 cases. Am J Obstet Gynecol 2022; 227:755.e1-755.e10. [PMID: 35667417 DOI: 10.1016/j.ajog.2022.05.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/16/2022] [Accepted: 05/30/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND In monochorionic twin pregnancies, the fetuses share a single placenta. When this placenta is unequally shared, a discordant antenatal growth pattern ensues resulting in high rates of perinatal morbidity and mortality. Understanding placental pathophysiology is paramount in devising feasible antenatal management strategies. Unequal placental sharing is not the sole determinant of birthweight discordance as there is no one-to-one relationship with placental share discordance. Placental angioarchitecture, especially the presence of large bidirectional anastomoses, is thought to affect this relationship by allowing for a compensatory intertwin blood flow. OBJECTIVE This study aimed to assess whether placental angioarchitecture can affect birthweight discordance in live-born monochorionic twins, the aim of our study was 2-fold: (1) to assess the relationship between birthweight discordance and placental share discordance and (2) to examine to what extent large bidirectional anastomoses can compensate for the effect of unequal placental sharing on birthweight discordance, with a subgroup analysis for umbilical artery Doppler flow patterns in cases with a birthweight discordance of ≥20%. STUDY DESIGN This was a retrospective cohort study that included monochorionic twin pregnancies observed in our center between March 2002 and June 2021, in which twins with a birthweight discordance of ≥20% were classified according to umbilical artery Doppler flow patterns of the smaller twin. We excluded cases with twin-twin transfusion syndrome and twin anemia polycythemia sequence. Monochorionic placentas of live-born twins were injected with dye, and images were saved for computer measurements of placental sharing and the diameter of anastomoses. Univariate linear regressions of the relationship between placental share discordance and birthweight discordance (both calculated as larger weight or share-smaller weight or share/larger weight or share×100%) and the relationship between arterioarterial and venovenous diameters and birthweight ratio/placental territory ratio were performed. RESULTS A total of 449 placentas were included in the analysis. Placental share discordance was positively correlated with birthweight discordance (β coefficient, 0.325; 95% confidence interval, 0.254-0.397; P<.0001). The arterioarterial diameter was negatively correlated with birthweight ratio/placental territory ratio (β coefficient, -0.041; 95% confidence interval, -0.059 to -0.023; P<.0001), meaning that an increase in arterioarterial diameter leads to less birthweight discordance than expected for the amount of placental share discordance. There was no relationship between venovenous diameter and birthweight ratio/placental territory ratio (β coefficient, -0.007; 95% confidence interval, -0.027 to 0.012; P=.473). CONCLUSION Birthweight discordance in monochorionic twins was strongly associated with placental share discordance. Large arterioarterial anastomoses can mitigate the effect of unequal placental sharing.
Collapse
Affiliation(s)
- Sophie G Groene
- Division of Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands; Division of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | - Femke Slaghekke
- Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique C Haak
- Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Bastiaan T Heijmans
- Division of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeanine M M van Klink
- Division of Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Arno A W Roest
- Division of Pediatric Cardiology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Enrico Lopriore
- Division of Neonatology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
19
|
Jordan B, Bernard L, Segel S, Go M, Schilling D, McEvoy C. Premature monochorionic monoamniotic twins have lower lung compliance at birth than matched dichorionic diamniotic twins. J Neonatal Perinatal Med 2022; 16:87-92. [PMID: 36314220 PMCID: PMC10168699 DOI: 10.3233/npm-221113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Premature infants are born with immature lungs that demonstrate abnormal pulmonary function with differences in passive respiratory system compliance and resistance, and functional residual capacity. To our knowledge, no studies have evaluated differences in neonatal pulmonary function based on the type of twin gestation, or chorionicity. Given the effect of chorionicity on outcomes, we aimed to study the effect of twin type, monochorionic monoamniotic (MCMA) vs dichorionic diamniotic (DCDA), on neonatal early pulmonary function tests. METHODS: In this prospective cohort study, 5 sets of DCDA twins were matched to 5 sets of MCMA twins on gestational age at delivery, latency from antenatal corticosteroid exposure, birthweight, race and gender. Mean values were compared for passive respiratory system compliance and resistance, functional residual capacity, and tidal volume. RESULTS: MCMA infants had a significantly lower compliance (0.64 vs 1.25 mL/cm H2O /kg; p = 0.0001) and significantly higher resistance (0.130 vs 0.087 cm H2O /mL/sec; p = 0.0003) than DCDA infants. Functional residual capacity was lower for MCMA than DCDA infants (17.5 vs 23.4 mL/kg; p = 0.17). Further, 80% of MCMA infants required intubation for surfactant administration compared to 20% of DCDA infants, indicating the clinical significance of these objective measures. CONCLUSIONS: Due to the matched case-control design, causality cannot be established. However, we speculate that these differences in lung function may derive from differential exposure to preterm labor and endogenous maternal corticosteroid exposure. Further study is necessary to establish the true causal relationship.
Collapse
Affiliation(s)
- B.K. Jordan
- Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, Oregon, USA
| | - L. Bernard
- Asante Physician Partners, Medford, Oregon, USA
| | - S. Segel
- Peace Health Southwest Medical Center, Vancouver, Washington, USA
| | - M.D. Go
- Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, Oregon, USA
| | - D. Schilling
- Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, Oregon, USA
| | - C.T. McEvoy
- Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
20
|
Mangiza M, Ehret DEY, Edwards EM, Rhoda N, Tooke L. Morbidity and mortality in small for gestational age very preterm infants in a middle-income country. Front Pediatr 2022; 10:915796. [PMID: 36016879 PMCID: PMC9396138 DOI: 10.3389/fped.2022.915796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/13/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE To evaluate the impact of small for gestational age (SGA) on outcomes of very preterm infants at Groote Schuur Hospital (GSH), Cape Town, South Africa. STUDY DESIGN Data were obtained from the Vermont Oxford Network (VON) GSH database from 2012 to 2018. The study is a secondary analysis of prospectively collected observational data. Fenton growth charts were used to define SGA as birth weight < 10th centile for gestational age. RESULTS Mortality [28.9% vs. 18.5%, adjusted risk ratio (aRR) 2.1, 95% confidence interval (CI) 1.6-2.7], bronchopulmonary dysplasia (BPD; 14% vs. 4.5%, aRR 3.7, 95% CI 2.3-6.1), and late-onset sepsis (LOS; 16.7% vs. 9.6%, aRR 2.3, 95% CI 1.6-3.3) were higher in the SGA than in the non-SGA group. CONCLUSION Small for gestational age infants have a higher risk of mortality and morbidity among very preterm infants at GSH. This may be useful for counseling and perinatal management.
Collapse
Affiliation(s)
- Marcia Mangiza
- Groote Schuur Hospital, Department of Paediatrics, University of Cape Town, Cape Town, South Africa
| | - Danielle E Y Ehret
- Vermont Oxford Network, Burlington, VT, United States.,Department of Paediatrics, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Erika M Edwards
- Vermont Oxford Network, Burlington, VT, United States.,Department of Paediatrics, Larner College of Medicine, University of Vermont, Burlington, VT, United States.,Department of Mathematics and Statistics, University of Vermont, Burlington, VT, United States
| | - Natasha Rhoda
- Groote Schuur Hospital, Department of Paediatrics, University of Cape Town, Cape Town, South Africa
| | - Lloyd Tooke
- Groote Schuur Hospital, Department of Paediatrics, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
21
|
Bauer SE, Vanderpool CPB, Ren C, Cristea AI. Nutrition and growth in infants with established bronchopulmonary dysplasia. Pediatr Pulmonol 2021; 56:3557-3562. [PMID: 34415681 DOI: 10.1002/ppul.25638] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 07/06/2021] [Accepted: 08/17/2021] [Indexed: 12/23/2022]
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common late morbidity of preterm birth. Ongoing clinical care and research have largely focused on the pathogenesis and prevention of BPD in preterm infants. However, preterm infants who develop BPD have significant medical needs that persist throughout their neonatal intensive care unit course and continue post-discharge, including those associated with growth and nutrition. The objective of this manuscript was to provide a review on nutrition and growth in infants with established BPD after discharge from the hospital and to identify the knowledge and research gaps to provide direction for future studies.
Collapse
Affiliation(s)
- Sarah E Bauer
- Department of Pediatrics, Indiana University, Indianapolis, Indiana, USA
| | | | - Clement Ren
- Division of Pulmonary and Sleep Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Aura Ioana Cristea
- Department of Pediatrics, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|