1
|
Wei S, Xiao M, Hu Y, Chang Y, Wang F, Liu L. Circ_19038 and lnc-AK016022 synergistically regulate Sirt1 to promote remyelination and alleviate white matter injury in preterm mice. Arch Biochem Biophys 2024; 760:110108. [PMID: 39084281 DOI: 10.1016/j.abb.2024.110108] [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: 04/21/2024] [Revised: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Maternal inflammation can lead to premature birth and fetal brain damage. CircRNA_19038 and lncRNA-AK016022 have been shown to be significantly reduced in brain tissues of preterm mice, while whether they are involved in the regulation of preterm white matter injury remains to be explored. Pregnant mice were intraperitoneally injected with lipopolysaccharide (LPS) to establish a preterm brain injury model. Healthy mice born at term served as controls. Lentivirus-mediated circ_19038 overexpression vector (LV-circ_19038), LV-lnc-AK016022, LV-Sirt1 and LV-sh-Sirt1 were administered to preterm mice through the ventricles. The expression levels of circ_19038, lnc-AK016022 and Sirt1 in the brain tissues of preterm mice were significantly lower than those of full-term healthy mice, and circ_19038 and lnc-AK016022 were co-localized in the brain tissues. Upregulation of circ_19038 or/and lnc-AK016022 promoted remyelination and alleviated white matter structural damage, neuroinflammation, and long-term cognitive and motor deficits in preterm mice, and the combined effect of circ_19038 and lnc-AK016022 showed better results. Primary mouse neuronal cells were isolated to investigate the regulatory effects of circ_19038 and lnc-AK016022 on Sirt1. Circ_19038 and lnc-AK016022 jointly promoted the expression of Sirt1 by adsorbing miR-1b and miR-328, respectively. Moreover, silencing Sirt1 antagonized the beneficial effects of circ_19038 or/and lnc-AK016022 on brain white matter injury in preterm mice. In conclusion, circ_19038 and lnc-AK016022 synergistically regulated Sirt1 expression to promote remyelination and alleviate white matter injury in preterm mice.
Collapse
Affiliation(s)
- Simeng Wei
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Mi Xiao
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuxin Hu
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuzhu Chang
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Fanghui Wang
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li Liu
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
2
|
Kumar N, Bidkhori HR, Yawno T, Lim R, Inocencio IM. Therapeutic potential of extracellular vesicles derived from human amniotic epithelial cells for perinatal cerebral and pulmonary injury. Stem Cells Transl Med 2024; 13:711-723. [PMID: 38895873 PMCID: PMC11328935 DOI: 10.1093/stcltm/szae037] [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: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/21/2024] Open
Abstract
Lung and brain injury that occurs during the perinatal period leads to lifelong disability and is often driven and/or exacerbated by inflammation. Human amniotic epithelial cells (hAEC), which demonstrate immunomodulatory, anti-fibrotic, and regenerative capabilities, are being explored as a therapeutic candidate for perinatal injury. However, limitations regarding scalable manufacturing, storage, transport, and dose-related toxicity have impeded clinical translation. Isolated therapeutic extracellular vesicles (EVs) from stem and stem-like cells are thought to be key paracrine mediators of therapeutic efficacy. The unique characteristics of EVs suggest that they potentially circumvent the limitations of traditional cell-based therapies. However, given the novelty of EVs as a therapeutic, recommendations around ideal methods of production, isolation, storage, and delivery have not yet been created by regulatory agencies. In this concise review, we discuss the pertinence and limitations of cell-based therapeutics in perinatal medicine. We also review the preclinical evidence supporting the use of therapeutic EVs for perinatal therapy. Further, we summarize the arising considerations regarding adequate cell source, biodistribution, isolation and storage methods, and regulatory roadblocks for the development of therapeutic EVs.
Collapse
Affiliation(s)
- Naveen Kumar
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Hamid Reza Bidkhori
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton 3168, Victoria, Australia
| | - Rebecca Lim
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Ishmael Miguel Inocencio
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| |
Collapse
|
3
|
Chen C, Zhu S, Fu T, Chen Y, Chen D. The protective effects of Ferrostatin-1 against inflammation-induced preterm birth and fetal brain injury. J Reprod Immunol 2024; 164:104260. [PMID: 38761507 DOI: 10.1016/j.jri.2024.104260] [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: 01/28/2024] [Revised: 03/19/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
INTRODUCTION Recent studies have suggested the involvement of ferroptosis in preterm birth. Despite compelling evidence, the underlying mechanism remains unknown. This investigation aimed to determine the therapeutic effects of Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, in preterm birth and fetal brain injury. METHODS Human placenta samples and clinical data of participants were collected to ascertain whether placental ferroptosis was associated with preterm birth. Lipopolysaccharide (LPS)-induced preterm birth mouse model was used to examine the protective effects of Fer-1 on preterm birth. Fetal brain tissues and offspring mice at 5 and 8 weeks were studied to determine the effects of Fer-1 on the cognitive function of offspring. RESULTS We examined the mechanism of spontaneous preterm birth and discovered that placental ferroptosis was associated with preterm birth. Fer-1 inhibited preterm birth by ameliorating placental ferroptosis and maternal inflammation, thus improving LPS-induced intrauterine inflammation to maintain pregnancy. Antenatal administration of Fer-1 prevented LPS-induced fetal brain damage in the acute phase and improved long-term neurodevelopmental impairments by improving placental neuroendocrine signaling and maintaining placental function. CONCLUSION Fer-1 inhibited preterm birth and fetal brain injury by inhibiting maternal inflammation and improving placental function. Our findings provide a novel therapeutic strategy for preterm birth.
Collapse
Affiliation(s)
- Chaolu Chen
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Shuaiying Zhu
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Tiantian Fu
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Yanmin Chen
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Danqing Chen
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China.
| |
Collapse
|
4
|
Asaka MK, Nishimura T, Kuwabara H, Itoh H, Takahashi N, Tsuchiya KJ. Interleukin-23 levels in umbilical cord blood are associated with neurodevelopmental trajectories in infancy. PLoS One 2024; 19:e0301982. [PMID: 38593153 PMCID: PMC11003674 DOI: 10.1371/journal.pone.0301982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Abstract
Our previous study, which aimed to understand the early neurodevelopmental trajectories of children with and without neurodevelopmental disorders, identified five classes of early neurodevelopmental trajectories, categorized as high normal, normal, low normal, delayed, and markedly delayed. This investigation involved measurement using the Mullen Scale of Early Learning in a representative sample of Japanese infants followed up from the age of 0 to 2 years (Nishimura et al., 2016). In the present study, we investigated the potential association between cytokine concentrations in umbilical cord serum with any of the five classes of neurodevelopmental trajectories previously assigned, as follows: high normal (N = 85, 13.0%), normal (N = 322, 49.1%), low normal (N = 137, 20.9%), delayed (N = 87, 13.3%), and markedly delayed (N = 25, 3.8%) in infancy. Decreased interleukin (IL)-23 levels in the cord blood were associated with the markedly delayed class, independent of potential confounders (odds ratio, 0.44; 95%confidence interval: 0.26-0.73). Furthermore, IL-23 levels decreased as the developmental trajectory became more delayed, demonstrating that IL-23 plays an important role in development, and is useful for predicting the developmental trajectory at birth.
Collapse
Affiliation(s)
- Machiko K. Asaka
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
| | - Tomoko Nishimura
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
- Research Centre for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hitoshi Kuwabara
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Psychiatry, Saitama Medical University, Moroyama-chou, Saitama, Japan
| | - Hiroaki Itoh
- Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Nagahide Takahashi
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
- Department of Child and Adolescent Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji J. Tsuchiya
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
- Research Centre for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
5
|
Jiangxue H, Liling Y, Fang X, Shumei Y, Gengying L, Xuejun R, Yao Y, Chuan N, Jie Y, Zhuxiao R. Wnt5a-Flt1 activation contributes to preterm altered cerebral angiogenesis after prenatal inflammation. Pediatr Neonatol 2023; 64:528-537. [PMID: 36922327 DOI: 10.1016/j.pedneo.2023.01.002] [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: 10/06/2022] [Revised: 12/26/2022] [Accepted: 01/18/2023] [Indexed: 02/19/2023] Open
Abstract
OBJECTIVE Intraventricular hemorrhage (IVH) causes morbidity and mortality in preterm infants and prenatal exposure to inflammation contributes to brain injury. Moreover, prenatal exposure to severe inflammation increases the risk of IVH in preterm neonates. The current study investigated whether intrauterine exposure to inflammation affects cerebral angiogenesis and its underlying mechanisms. METHODS Wnt5a, flt1, and vascular endothelial growth factor (VEGF)-A levels in cord blood serum (stored in a bio-bank) of the enrolled patients were measured via enzyme-linked immunosorbent assay. A preterm prenatal inflammation exposure model was established in rats by intraperitoneal injection intraperitoneally during pregnancy. Angiogenesis of cerebral tissue was analyzed using immunohistochemistry. Wnt5a, flt1, and VEGF-A expression levels were measured via immunohistochemistry, immunofluorescence, or western blotting. The correlation between Wnt5a and flt1 expression and the cerebral vessel area was also analyzed. RESULTS The Wnt5a and flt1 levels in the cord blood serum were significantly higher in the amnionitis group than in the non-amnionitis group. The VEGF-A level in the cord blood serum was significantly lower in the amnionitis group. In the rat model, preterm rats in the prenatal inflammation group exhibited increased microglial cell infiltration and decreased vessel area and diameter in the cerebral tissue compared to the control group. Wnt5a was located in microglial cells, and Wnt5a and flt1 expression in brain tissue significantly increased after prenatal lipopolysaccharide (LPS) exposure. VEGF-A expression declined after prenatal LPS exposure. The cerebral vessel area was negatively correlated with Wnt5a and flt1 expression. CONCLUSION Disordered cerebral angiogenesis is associated with increased Wnt5a-Flt1 activation in microglial cells after exposure to intrauterine inflammation.
Collapse
Affiliation(s)
- Han Jiangxue
- Department of Neonatology, Guangdong Key Clinical Specialty, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yang Liling
- Department of Neonatology, Guangdong Key Clinical Specialty, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xu Fang
- Department of Neonatology, Guangdong Key Clinical Specialty, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yang Shumei
- Department of Neonatology, Guangdong Key Clinical Specialty, Guangdong Women and Children Hospital, Guangzhou, China
| | - Liu Gengying
- Department of Neonatology, Guangdong Key Clinical Specialty, Guangdong Women and Children Hospital, Guangzhou, China
| | - Ren Xuejun
- Dongguan Maternal and Children Hospital, Dong Guan, Guangdong, China
| | - Yao Yao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, China
| | - Nie Chuan
- Department of Neonatology, Guangdong Key Clinical Specialty, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yang Jie
- Department of Neonatology, Nanfang Hospital, Southern Medical University, China.
| | - Ren Zhuxiao
- Department of Neonatology, Guangdong Key Clinical Specialty, Guangdong Women and Children Hospital, Guangzhou, China.
| |
Collapse
|
6
|
Benny M, Bandstra ES, Saad AG, Lopez-Alberola R, Saigal G, Paidas MJ, Jayakumar AR, Duara S. Maternal SARS-CoV-2, Placental Changes and Brain Injury in 2 Neonates. Pediatrics 2023; 151:e2022058271. [PMID: 37021494 PMCID: PMC10467358 DOI: 10.1542/peds.2022-058271] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2023] [Indexed: 04/07/2023] Open
Abstract
Long-term neurodevelopmental sequelae are a potential concern in neonates following in utero exposure to severe acute respiratory syndrome coronavirus disease 2 (SARS-CoV-2). We report 2 neonates born to SARS-CoV-2 positive mothers, who displayed early-onset (day 1) seizures, acquired microcephaly, and significant developmental delay over time. Sequential MRI showed severe parenchymal atrophy and cystic encephalomalacia. At birth, neither infant was SARS-CoV-2 positive (nasopharyngeal swab, reverse transcription polymerase chain reaction), but both had detectable SARS-CoV-2 antibodies and increased blood inflammatory markers. Placentas from both mothers showed SARS-CoV-2-nucleocapsid protein and spike glycoprotein 1 in the syncytiotrophoblast, fetal vascular malperfusion, and significantly increased inflammatory and oxidative stress markers pyrin domain containing 1 protein, macrophage inflammatory protein 1 βη, stromal cell-derived factor 1, interleukin 13, and interleukin 10, whereas human chorionic gonadotropin was markedly decreased. One infant (case 1) experienced sudden unexpected infant death at 13 months of age. The deceased infant's brain showed evidence of SARS-CoV-2 by immunofluorescence, with colocalization of the nucleocapsid protein and spike glycoprotein around the nucleus as well as within the cytoplasm. The constellation of clinical findings, placental pathology, and immunohistochemical changes strongly suggests that second-trimester maternal SARS-CoV-2 infection with placentitis triggered an inflammatory response and oxidative stress injury to the fetoplacental unit that affected the fetal brain. The demonstration of SARS-CoV-2 in the deceased infant's brain also raises the possibility that SARS-CoV-2 infection of the fetal brain directly contributed to ongoing brain injury. In both infants, the neurologic findings at birth mimicked the presentation of hypoxic-ischemic encephalopathy of newborn and neurologic sequelae progressed well beyond the neonatal period.
Collapse
Affiliation(s)
- Merline Benny
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| | - Emmalee S. Bandstra
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| | - Ali G. Saad
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
| | - Roberto Lopez-Alberola
- Division of Child Neurology Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida
| | - Gaurav Saigal
- Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Michael J. Paidas
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - Arumugam R. Jayakumar
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - Shahnaz Duara
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
7
|
Dong X, Zhou A. Associations of maternal pre-pregnancy body mass index and gestational weight gain with risk of offspring neurodevelopment at 2 years: A Chinese birth cohort study. Front Pediatr 2023; 11:1165743. [PMID: 37144148 PMCID: PMC10151668 DOI: 10.3389/fped.2023.1165743] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/21/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction In recent decades, there has been a surge in both obesity and developmental impairments. Only a few research have looked at the relationship between gestational weight growth and pre-pregnancy BMI in mothers and the neurobehavioral development of their infants. The current research investigates the associations among maternal pre-pregnancy BMI, GWG, and the risk of child neural development at 2 years of age depending on a Chinese birth prospective study. Methods The study population was 3,115 mother-infant pairs were registered in the Wuhan Health Baby cohort between September 2013 and October 2018, and data from this cohort was used in this investigation. The Chinese classification was used to group maternal BMI before conception. Based on the 2019 Life Cycle Project-Maternal Obesity and Childhood Outcomes Study Group, categories for GWG were created. The outcome was an assessment of child neural development at age 2 which was measured by employing a Chinese translation of the Bayley scales (BSID-CR). The multivariate regression models were used to calculate the beta (β) coefficients and 95% confidence intervals (CIs) for estimating the associations between continuous Bayley scores and maternal pre-pregnancy BMI categories, as same as in GWG categories. Results Infants of overweight and obese moms exhibited lower MDI scores than those of mothers with normal pre-pregnancy BMI (β = -2.510, 95%CI = -4.821 to -0.200) in the entire sample. Meanwhile, we find among the normal pre-pregnancy BMI mothers, infants of inadequate GWG mothers had lower MDI scores (β = -3.952, 95%CI = -7.809 to -0.094) compared with the referenced adequate GWG mothers, as well as the infants of excessive GWG mothers among the underweight pre-pregnancy BMI mothers (β = -5.173, 95%CI = -9.803 to -0.543). The PDI scores of the infants were not affected by the maternal pre-pregnancy BMI or GWG. Conclusion For Chinese babies aged 2 in this nationally representative sample, aberrant pre-pregnancy BMI and GWG can impair infants' mental development, but not psychomotor development. Such results are significant given the incidence of overweight and obesity as well as the long-term effects of early brain development. In this study we found optimal GWG recommendations proposed by 2019 Life Cycle Project-Maternal Obesity and Childhood Outcomes Study Group were more suitable for Chinese women than 2009 Institute of Medicine(IOM) guidelines. Additionally, women should be given general advice on how to achieve their ideal pre-pregnancy BMI and GWG.
Collapse
Affiliation(s)
- Xiaohan Dong
- Department of Obstetrics and Gynecology, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Aifen Zhou
- Department of Obstetrics and Gynecology, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Institute of Maternal and Child Health, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Correspondence: Aifen Zhou
| |
Collapse
|
8
|
Quinte GC, Munhoz TN, Matijasevich A, Santos IS. Maternal pre-pregnancy body mass index and mental health problems in early adolescents from the 2004 Pelotas birth cohort. Sci Rep 2022; 12:14437. [PMID: 36002596 PMCID: PMC9402554 DOI: 10.1038/s41598-022-18032-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
Possible mechanisms by which maternal pre-pregnancy body mass index (BMI) programs offspring mental disorders in late childhood are not fully clarified. To assess the association between maternal BMI and mental health problems at 11 years old, we used data from the 2004 Pelotas birth cohort which comprised 4231 newborns. Maternal pre-pregnancy BMI was analyzed as underweight (< 18.5), normal (18.5-24.9), overweight (25.0-29.9), and obesity (≥ 30.0). Mental health problems were assessed at the child's age of 11 years by the Strengths and Difficulties Questionnaire, total difficulties score and subscale scores (emotional symptoms, conduct problems, hyperactivity/inattention, and peer relationship problems), dichotomized into normal/borderline and abnormal category. The associations between maternal pre-pregnancy BMI and mental health problems in the whole sample and stratified by sex were estimated using crude and adjusted linear and logistic regression. Both linear and logistic regression showed that pre-pregnancy weight was associated with mental problems in early adolescents. Maternal pre-pregnancy obesity was associated with increased odds of total difficulty score among the whole sample. Boys whose mothers were pre-pregnancy overweight had higher odds of conduct problems (OR = 1.56; 95% CI: 1.13, 1.95), when compared to children of normal pre-pregnancy weight mothers, even after adjustments. Maternal pre-pregnancy obesity was associated with mental health problems in early adolescents; specifically, pre-pregnancy overweight increased the risk for conduct problems in 11 years old boys.
Collapse
Affiliation(s)
- Gabriela Callo Quinte
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Rua Marechal Deodoro, 1160 - 3° Piso, Bairro Centro, Caixa Postal 464, Pelotas, RS, Cep: 96020-220, Brazil.
| | - Tiago N Munhoz
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Rua Marechal Deodoro, 1160 - 3° Piso, Bairro Centro, Caixa Postal 464, Pelotas, RS, Cep: 96020-220, Brazil.,School of Psychology, Federal University of Pelotas, Pelotas, Brazil
| | - Alicia Matijasevich
- Departamento de Medicina Preventiva, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ina S Santos
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Rua Marechal Deodoro, 1160 - 3° Piso, Bairro Centro, Caixa Postal 464, Pelotas, RS, Cep: 96020-220, Brazil
| |
Collapse
|
9
|
Kirschen GW, Panda S, Burd I. Congenital Infection Influence on Early Brain Development Through the Gut-Brain Axis. Front Neurosci 2022; 16:894955. [PMID: 35844234 PMCID: PMC9280077 DOI: 10.3389/fnins.2022.894955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/23/2022] [Indexed: 11/21/2022] Open
Abstract
The mechanisms by which various pathogens cause congenital infections have been studied extensively, aiding in the understanding of the detrimental effects these infections can have on fetal/neonatal neurological development. Recent studies have focused on the gut-brain axis as pivotal in neurodevelopment, with congenital infections causing substantial disruptions. There remains controversy surrounding the purported sterility of the placenta as well as concerns regarding the effects of exposure to antibiotics used during pregnancy on neonatal microbiome development and how early exposure to microbes or antibiotics can shape the gut-brain axis. Long-term neurodevelopmental consequences, such as autism spectrum disorder, attention deficit hyperactivity disorder, and cerebral palsy, may be attributable, in part, to early life infection and changes in the immature gut microbiome. The goal of this review is thus to critically evaluate the current evidence related to early life infection affecting neurodevelopment through the gut-brain axis.
Collapse
Affiliation(s)
- Gregory W. Kirschen
- Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD, United States
- Integrated Center for Fetal Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Snigdha Panda
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Irina Burd
- Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD, United States
- Integrated Center for Fetal Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
10
|
Wang B, Zou L, Li M, Zhou L. Astrocyte: A Foe or a Friend in Intellectual Disability-Related Diseases. Front Synaptic Neurosci 2022; 14:877928. [PMID: 35812794 PMCID: PMC9259964 DOI: 10.3389/fnsyn.2022.877928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022] Open
Abstract
Intellectual disabilities are a type of neurodevelopmental disease caused by neurological dysfunction. Their incidence is largely associated with neural development. Astrocytes are the most widely distributed cells in the mammalian brain. Previous studies have reported that astrocytes only supported and separated the neurons in the brain. However, recent studies have found that they also play an important role in neural development. Understanding the astrocyte mechanism in intellectual development disorder-related diseases will help provide new therapeutic targets for the treatment of intellectual disability-related diseases. This mini-review introduced the association between astrocyte and intellectual disabilities. Furthermore, recent advances in genetic and environmental factors causing intellectual disability and different pharmaceutical effects of intellectual disability-related drugs on astrocytes have been summarised. Finally, we discussed future perspectives of astrocyte-based therapy for intellectual disability.
Collapse
Affiliation(s)
| | | | | | - Liang Zhou
- *Correspondence: Liang Zhou, , orcid.org/0000-0003-0820-1520
| |
Collapse
|
11
|
Ghanavatinejad A, Rashidi N, Mirahmadian M, Rezania S, Mosalaei M, Ghasemi J, Zarnani AH. Vitamin D3 Controls TLR4- and TLR2-Mediated Inflammatory Responses of Endometrial Cells. Gynecol Obstet Invest 2021; 86:139-148. [PMID: 33540416 DOI: 10.1159/000513590] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 11/26/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Vitamin D has potent immunoregulatory features and modulates innate and adaptive immune responses. There is a significant association between intrauterine infection-associated inflammatory responses and pregnancy complications such as abortion and preterm labor. Here, we investigated how 1,25 (OH)2 D3 could modulate inflammatory responses of endometrial cells. DESIGN This is an in vitro experimental study. Endometrial stromal cells (ESCs) and whole endometrial cells (WECs) were collected from 15 apparently normal women, and the immunomodulatory effects of 1,25 (OH)2 D3 on lipopolysaccharide (LPS)- or lipoteichoic acid (LTA)-treated ESCs and WECs were investigated. Participants/Materials, Setting, and Methods: Women with no history of abortion, infertility, endometriosis, or sign of vaginal infection were enrolled in this study. Endometrial samples were collected by gynecologists using a Pipelle pipette in the proliferative phase of the menstrual cycle. WECs and ESCs were collected and treated with either LPS or LTA. The levels of IL-6, IL-8, and TNF-α in culture supernatants were quantified using the ELISA technique. TLR2, TLR4, and MyD88 expressions were assessed by RT-qPCR. TLR4 expression at the protein level was studied by the Western blot technique. RESULTS 1,25 Dihydroxycholecalciferol (1,25 (OH)2 D3) significantly reduced TNF-α production in LPS-activated ESCs and TNF-α and IL-6 production by LTA-stimulated WECs. In contrast, 1,25 (OH)2 D3 pretreatment increased the production of IL-8 by LPS- and LTA-stimulated endometrial cells. 1,25 (OH)2 D3 pretreatment markedly reduced LPS-induced TLR4 protein expression by ESCs. LPS treatment of ESCs significantly induced MyD88 gene expression. This effect was reversed when these cells were pretreated with 1,25 (OH)2 D3 before stimulation with LPS. LIMITATIONS Because of the small size of samples, doing experiments all together on some samples was not feasible. Confirmation of the results obtained here needs well-designed in vivo studies. CONCLUSIONS 1,25 (OH)2 D3 is an immunomodulatory molecule essential for maintaining endometrial immune homeostasis by controlling potentially harmful inflammatory responses associated with female reproductive tract infections.
Collapse
Affiliation(s)
- Alireza Ghanavatinejad
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nesa Rashidi
- Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran
| | - Mahroo Mirahmadian
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Simin Rezania
- Biophysics Institute, Medical University of Graz, Graz, Austria
| | - Mahdokht Mosalaei
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Jamileh Ghasemi
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran, .,Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran, .,Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran,
| |
Collapse
|
12
|
Matar R, Alrahmani L, Monzer N, Debiane LG, Berbari E, Fares J, Fitzpatrick F, Murad MH. Clinical Presentation and Outcomes of Pregnant Women With Coronavirus Disease 2019: A Systematic Review and Meta-analysis. Clin Infect Dis 2021; 72:521-533. [PMID: 32575114 PMCID: PMC7337697 DOI: 10.1093/cid/ciaa828] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Descriptions of coronavirus disease 2019 (COVID-19) have focused on the nonpregnant adult population. This study aims to describe the clinical characteristics and perinatal outcomes of COVID-19 in pregnancy. METHODS We searched databases from December 2019 to 30 April 2020. Eligible studies reported clinical characteristics, radiological findings, and/or laboratory testing of pregnant women during infection. Data were pooled across studies using a random-effects model. RESULTS Twenty-four studies (136 women) were included. The most common symptoms were fever (62.9%) and cough (36.8%). Laboratory findings included elevated C-reactive protein (57%) and lymphocytopenia (50%). Ground-glass opacity was the most common radiological finding (81.7%). Preterm birth rate was 37.7% and cesarean delivery rate was 76%. There was 1 maternal death. There were 2 fetal COVID-19 cases. CONCLUSIONS The clinical picture in pregnant women with COVID-19 did not differ from the nonpregnant population; however, the rate of preterm birth and cesarean delivery are considerably higher than international averages.
Collapse
Affiliation(s)
- Reem Matar
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Layan Alrahmani
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Loyola University Medical Center, Chicago, Illinois, USA
| | - Nasser Monzer
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Labib G Debiane
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Henry Ford, Detroit, Michigan, USA
| | - Elie Berbari
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jawad Fares
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois, USA
| | - Fidelma Fitzpatrick
- Department of Microbiology, Beaumont Hospital, Dublin, Ireland
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mohammad H Murad
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
13
|
Ginsberg Y, Gutzeit O, Hadad S, Divon MY, Khatib N, Fainaru O, Weiner Z, Beloosesky R. Maternal Progesterone Treatment Reduces Maternal Inflammation-Induced Fetal Brain Injury in a Mouse Model of Preterm Birth. Reprod Sci 2021; 28:166-176. [PMID: 32833191 DOI: 10.1007/s43032-020-00272-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/22/2020] [Indexed: 01/08/2023]
Abstract
Maternal natural vaginal progesterone (nVP) administration has been shown to reduce the risk of preterm birth (PTB). The largest randomized trial of nVP for PTB (OPPTIMUM) noted a sonographic reduction in neonatal brain injury following nVP treatment. We investigated the neuroinflammatory protective effect of maternal nVP in a mouse model for maternal inflammation. Pregnant mice (n = 24) were randomized to nVP (1 mg/day) or vehicle from days 13-16 of gestation. At days 15 and 16, lipopolysaccharide (30 μg) or saline were administered. Mice were sacrificed 4 h following the last injection. Fetal brains and placentas were collected. Levels of NF-κB, nNOS, IL-6, and TNFα were determined by Western blot. Maternal lipopolysaccharide significantly increased fetal brain levels of IL-6 (0.33 ± 0.02 vs. 0.11 ± 0.01 u), TNFα (0.3 ± 0.02 vs. 0.10 ± 0.01 u), NF-κB (0.32 ± 0.01 vs. 0.17 ± 0.01 u), and nNOS (0.24 ± 0.04 vs. 0.08 ± 0.01 u), and reduced the total glutathione levels (0.014 ± 0.001 vs. 0.026 ± 0.001 pmol/μl; p < 0.01) compared with control. Maternal nVP significantly reduced fetal brain levels of IL-6 (0.14 ± 0.01 vs. 0.33 ± 0.02 u), TNFα (0.2 ± 0.06 vs. 0.3 ± 0.02 u), NF-κB (0.16 ± 0.01 vs 0.32 ± 0.01 u), and nNOS (0.14 ± 0.01 vs 0.24 ± 0.04 u), and prevented the reduction of fetal brain total glutathione levels (0.022 ± 0.001 vs. 0.014 ± 0.001 pmol/μl; p < 0.01) to levels similar to controls. A similar pattern was demonstrated in the placenta. Maternal nVP for PTB may protect the fetal brain from inflammation-induced brain injury by inhibiting specific inflammatory and oxidative pathways in both brain and placenta.
Collapse
Affiliation(s)
- Yuval Ginsberg
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel.
| | - Ola Gutzeit
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Salim Hadad
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Michael Y Divon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Lenox Hill Hospital, Northwell Health, New York City, NY, USA
| | - Nizar Khatib
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Ofer Fainaru
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Ron Beloosesky
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| |
Collapse
|
14
|
Koehn LM, Huang Y, Habgood MD, Kysenius K, Crouch PJ, Dziegielewska KM, Saunders NR. Effects of paracetamol (acetaminophen) on gene expression and permeability properties of the rat placenta and fetal brain. F1000Res 2020; 9:573. [PMID: 32934805 PMCID: PMC7477648 DOI: 10.12688/f1000research.24119.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2020] [Indexed: 03/30/2024] Open
Abstract
Background: Paracetamol (acetaminophen) is widely used in pregnancy and generally regarded as "safe" by regulatory authorities. Methods: Clinically relevant doses of paracetamol were administered intraperitoneally to pregnant rats twice daily from embryonic day E15 to 19 (chronic) or as a single dose at E19 (acute). Control samples were from un-treated age-matched animals. At E19, rats were anaesthetised, administered a final paracetamol dose, uteruses were opened and fetuses exposed for sample collection. For RNA sequencing, placentas and fetal brains were removed and flash frozen. Fetal and maternal plasma and cerebrospinal fluid were assayed for ⍺-fetoprotein and interleukin 1β (IL1β). Brains were fixed and examined (immunohistochemistry) for plasma protein distribution. Placental permeability to a small molecule ( 14C-sucrose) was tested by injection into either mother or individual fetuses; fetal and maternal blood was sampled at regular intervals to 90 minutes. Results: RNA sequencing revealed a large number of genes up- or down-regulated in placentas from acutely or chronically treated animals compared to controls. Most notable was down-regulation of three acute phase plasma proteins (⍺-fetoprotein, transferrin, transthyretin) in acute and especially chronic experiments and marked up-regulation of immune-related genes, particularly cytokines, again especially in chronically treated dams. IL1β increased in plasma of most fetuses from treated dams but to variable levels and no IL1β was detectable in plasma of control fetuses or any of the dams. Increased placental permeability appeared to be only from fetus to mother for both 14C-sucrose and ⍺-fetoprotein, but not in the reverse direction. In the fetal brain, gene regulatory changes were less prominent than in the placenta of treated fetuses and did not involve inflammatory-related genes; there was no evidence of increased blood-brain barrier permeability. Conclusion: Results suggest that paracetamol may induce an immune-inflammatory-like response in placenta and more caution should be exercised in use of paracetamol in pregnancy.
Collapse
Affiliation(s)
- Liam M. Koehn
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yifan Huang
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mark D Habgood
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Kai Kysenius
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Peter J. Crouch
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | - Norman R Saunders
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
15
|
Koehn LM, Huang Y, Habgood MD, Kysenius K, Crouch PJ, Dziegielewska KM, Saunders NR. Effects of paracetamol (acetaminophen) on gene expression and permeability properties of the rat placenta and fetal brain. F1000Res 2020; 9:573. [PMID: 32934805 PMCID: PMC7477648 DOI: 10.12688/f1000research.24119.2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2020] [Indexed: 01/10/2023] Open
Abstract
Background: Paracetamol (acetaminophen) is widely used in pregnancy and generally regarded as "safe" by regulatory authorities. Methods: Clinically relevant doses of paracetamol were administered intraperitoneally to pregnant rats twice daily from embryonic day E15 to 19 (chronic) or as a single dose at E19 (acute). Control samples were from un-treated age-matched animals. At E19, rats were anaesthetised, administered a final paracetamol dose, uteruses were opened and fetuses exposed for sample collection. For RNA sequencing, placentas and fetal brains were removed and flash frozen. Fetal and maternal plasma and cerebrospinal fluid were assayed for α-fetoprotein and interleukin 1β (IL1β). Brains were fixed and examined (immunohistochemistry) for plasma protein distribution. Placental permeability to a small molecule ( 14C-sucrose) was tested by injection into either mother or individual fetuses; fetal and maternal blood was sampled at regular intervals to 90 minutes. Results: RNA sequencing revealed a large number of genes up- or down-regulated in placentas from acutely or chronically treated animals compared to controls. Most notable was down-regulation of three acute phase plasma proteins (α-fetoprotein, transferrin, transthyretin) in acute and especially chronic experiments and marked up-regulation of immune-related genes, particularly cytokines, again especially in chronically treated dams. IL1β increased in plasma of most fetuses from treated dams but to variable levels and no IL1β was detectable in plasma of control fetuses or any of the dams. Increased placental permeability appeared to be only from fetus to mother for both 14C-sucrose and α-fetoprotein, but not in the reverse direction. In the fetal brain, gene regulatory changes were less prominent than in the placenta of treated fetuses and did not involve inflammatory-related genes; there was no evidence of increased blood-brain barrier permeability. Conclusion: Results suggest that paracetamol may induce an immune-inflammatory-like response in placenta and more caution should be exercised in use of paracetamol in pregnancy.
Collapse
Affiliation(s)
- Liam M. Koehn
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yifan Huang
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mark D Habgood
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Kai Kysenius
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Peter J. Crouch
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | - Norman R Saunders
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
16
|
Dong J, Lei J, Elsayed NA, Lee JY, Shin N, Na Q, Chudnovets A, Jia B, Wang X, Burd I. The effect of intrauterine inflammation on mTOR signaling in mouse fetal brain. Dev Neurobiol 2020; 80:149-159. [PMID: 32333505 DOI: 10.1002/dneu.22755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/24/2019] [Accepted: 04/20/2020] [Indexed: 11/05/2022]
Abstract
Fetuses exposed to an inflammatory environment are predisposed to long-term adverse neurological outcomes. However, the mechanism by which intrauterine inflammation (IUI) is responsible for abnormal fetal brain development is not fully understood. The mechanistic target of rapamycin (mTOR) signaling pathway is closely associated with fetal brain development. We hypothesized that mTOR signaling might be involved in fetal brain injury and malformation when fetuses are exposed to the IUI environment. A well-established IUI model was utilized by intrauterine injection of lipopolysaccharide (LPS) to explore the effect of IUI on mTOR signaling in mouse fetal brains. We found that microglia activation in LPS fetal brains was increased, as demonstrated by elevated Iba-1 protein level and immunofluorescence density. LPS fetal brains also showed reduced neuronal cell counts, decreased cell proliferation demonstrated by low Ki67-positive density, and elevated neuron apoptosis evidenced by high expression of cleaved Caspase 3. Furthermore, we found that mTOR signaling in LPS fetal brains was elevated at 2 hr after LPS treatment, declined at 6 hr and showed overall inhibition at 24 hr. In summary, our study revealed that LPS-induced IUI leads to increased activation of microglia cells, neuronal damage, and dynamic alterations in mTOR signaling in the mouse fetal brain. Our findings indicate that abnormal changes in mTOR signaling may underlie the development of future neurological complications in offspring exposed to prenatal IUI.
Collapse
Affiliation(s)
- Jie Dong
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Obstetrics and Gynecology, Reproductive Medical Center, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jun Lei
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nada A Elsayed
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ji Yeon Lee
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Na Shin
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Quan Na
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna Chudnovets
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bei Jia
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaohong Wang
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Irina Burd
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Relationship of prenatal maternal obesity and diabetes to offspring neurodevelopmental and psychiatric disorders: a narrative review. Int J Obes (Lond) 2020; 44:1981-2000. [PMID: 32494038 PMCID: PMC7508672 DOI: 10.1038/s41366-020-0609-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 04/20/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
Abstract
Obesity and diabetes is a worldwide public health problem among women of reproductive age. This narrative review highlights recent epidemiological studies regarding associations of maternal obesity and diabetes with neurodevelopmental and psychiatric disorders in offspring, and provides an overview of plausible underlying mechanisms and challenges for future human studies. A comprehensive search strategy selected terms that corresponded to the domains of interest (maternal obesity, different types of diabetes, offspring cognitive functions and neuropsychiatric disorders). The databases searched for articles published between January 2010 and April 2019 were PubMed, Web of Science and CINAHL. Evidence from epidemiological studies strongly suggests that maternal pre-pregnancy obesity is associated with increased risks for autism spectrum disorder, attention-deficit hyperactivity disorder and cognitive dysfunction with modest effect sizes, and that maternal diabetes is associated with the risk of the former two disorders. The influence of maternal obesity on other psychiatric disorders is less well studied, but there are reports of associations with increased risks for offspring depression, anxiety, schizophrenia and eating disorders, at modest effect sizes. It remains unclear whether these associations are due to intrauterine mechanisms or explained by confounding family-based sociodemographic, lifestyle and genetic factors. The plausible underlying mechanisms have been explored primarily in animal models, and are yet to be further investigated in human studies.
Collapse
|
18
|
Kong L, Nilsson IAK, Brismar K, Gissler M, Lavebratt C. Associations of Different Types of Maternal Diabetes and Body Mass Index With Offspring Psychiatric Disorders. JAMA Netw Open 2020; 3:e1920787. [PMID: 32031649 DOI: 10.1001/jamanetworkopen.2019.20787] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPORTANCE Maternal obesity, pregestational type 1 and 2 diabetes, and gestational diabetes have been reported to increase the risk of autism spectrum disorder and attention-deficit/hyperactivity disorder in the mothers' offspring. However, the associations of maternal diabetes disorders and body mass index jointly with psychiatric disorders among offspring are less well documented, especially for type 2 diabetes. OBJECTIVE To examine the associations of different types of maternal diabetes, separately and together with maternal obesity, with psychiatric disorders in the mothers' offspring. DESIGN, SETTING, AND PARTICIPANTS This population-based cohort study used data from nationwide registries in Finland encompassing all 649 043 live births occurring between 2004 and 2014. The study and data analysis were conducted from January 1, 2019, to July 5, 2019. EXPOSURES Maternal prepregnancy body mass index, insulin-treated pregestational diabetes, and pregestational type 2 diabetes and gestational diabetes without insulin treatment. MAIN OUTCOMES AND MEASURES Psychiatric diagnoses and prescription of psychotropic drugs among the mothers' offspring. Cox proportional hazards models were adjusted for birth year, sex, mode of delivery, maternal age, number of fetuses, parity, mother's country of birth, mother's marital status, maternal smoking, maternal psychiatric disorder, and maternal systemic inflammatory disease. RESULTS The mean (SD) age of mothers was 30.20 (5.37) years; 357 238 of 394 302 mothers (90.6%) were born in Finland. Of the 647 099 births studied, 4000 fetuses (0.62%) were exposed to maternal insulin-treated pregestational diabetes, 3724 (0.57%) were exposed to type 2 diabetes, and 98 242 (15.18%) were exposed to gestational diabetes; 34 892 offspring (5.39%) later received a diagnosis of a mild neurodevelopmental or psychiatric disorder. Non-insulin-treated type 2 diabetes in severely obese mothers, compared with normal-weight mothers without diabetes, was associated with psychiatric disorders in the offspring (hazard ratio, 1.97; 95% CI, 1.64-2.37), although with a lower effect size than that for severely obese mothers with insulin-treated pregestational diabetes (hazard ratio, 2.71; 95% CI, 2.03-3.61). The largest effect sizes were found for mood disorders, attention-deficit/hyperactivity disorder and conduct disorders, and autism. Gestational diabetes in severely obese mothers had a lower overall effect size (hazard ratio, 1.61; 95% CI, 1.50-1.72). Diabetes in normal-weight mothers was not associated with psychopathologic disorders in the offspring. CONCLUSIONS AND RELEVANCE Severe obesity in mothers with diabetes was associated with an increased overall risk for psychiatric disorders in their offspring. The risk was highest for those exposed to insulin-treated pregestational diabetes, followed by non-insulin-treated type 2 diabetes and gestational diabetes. These findings may have implications for managing pregnancies.
Collapse
Affiliation(s)
- Linghua Kong
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Translational Psychiatry Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ida A K Nilsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Translational Psychiatry Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Mika Gissler
- National Institute for Health and Welfare, Helsinki, Finland
- Division of Family Medicine, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Research Centre for Child Psychiatry, University of Turku, Turku, Finland
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Translational Psychiatry Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Hossin MZ, Björk J, Koupil I. Early-life social and health determinants of adult socioeconomic position: associations and trends across generations. J Epidemiol Community Health 2020; 74:412-420. [PMID: 31988239 PMCID: PMC7307663 DOI: 10.1136/jech-2019-213209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/23/2019] [Accepted: 01/11/2020] [Indexed: 11/23/2022]
Abstract
Background Social and biological circumstances at birth are established predictors of adult socioeconomic position (SEP). This study aims to assess the trends in these associations across two generations and examine the effects of parental early-life characteristics on descendants’ adult SEP. Methods We studied men and women born in the Uppsala University Hospital 1915–1929 (G1) and their offspring born 1932–1960 (G2). Data were collected in archives and routine registers. Adult SEP was assessed as an aggregate measure combining education and occupation. The exposures were family SEP, mother’s marital status, mother’s parity, mother’s age, standardised birth weight, gestational length and birth multiplicity. Linear regression was used to examine the associations across generations. Results The difference in adult SEP between low and high family SEP at birth was 15.8 (95% CI: 13.3 to 18.3) percentage points smaller in G2 compared with G1, although a considerable difference was still evident in G2. The associations of adult SEP with small birth weight for gestational age, post-term birth and high parity were stable between the generations: the generational differences in adjusted coefficients were 1.5 (95% CI: −1.1 to 4.1), 0.6 (–1.7 to 2.9) and 1.8 (–0.2 to 3.8) percentage points, respectively. The association between grandparental and grandchildren’s SEPs was largely explained by parental socioeconomic conditions. Father’s preterm birth was independently associated with offspring’s SEP. Conclusion The stability of the associations between early-life biological disadvantages and adult SEP and the persistent, although attenuated, association between early-life and adult SEPs necessitates increased policy attention to both social and health conditions at birth.
Collapse
Affiliation(s)
| | - Jonas Björk
- Department of Occupational and Environmental Medicine, Lunds Universitet, Lund, Sweden
| | - Ilona Koupil
- Department of Global Public Health, Karolinska Institute, Stockholm, Sweden.,Department of Public Health Sciences, Stockholm University, Stockholm, Sweden
| |
Collapse
|
20
|
Preterm birth subtypes, placental pathology findings, and risk of neurodevelopmental disabilities during childhood. Placenta 2019; 83:17-25. [PMID: 31477202 DOI: 10.1016/j.placenta.2019.06.374] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 05/24/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Preterm birth (PTB) and in-utero inflammation are recognized risk factors of neurodevelopmental disabilities (NDDs); however, their combined role in NDDs is unknown. We examined the independent and joint association of PTB and placental histological findings with the childhood risk of NDDs (overall and by subgroups including autism spectrum disorder (ASD) and ADHD). METHODS We analyzed data from the Boston Birth Cohort, where mother-infant pairs were enrolled at birth and followed from birth onwards. Birth outcomes, placental pathology and NDDs were obtained from electronic medical records. Placental pathology was categorized using a standardized classification system proposed by the Amsterdam Placental Workshop Group. RESULTS PTB (all, including spontaneous, medically indicated) was an independent risk factor for NDDs. Placental histological chorioamnionitis (CA) and PTB additively increased the odds of NDDs (aOR: 2.16, 95% CI: 1.37, 3.39), as well as ADHD (aOR: 2.75, 95% CI: 1.55, 4.90), other developmental disabilities (aOR: 1.96, 95% CI: 1.18, 3.25) and possibly ASD (aOR: 2.31, 95% CI: 0.99, 5.39). The above associations were more pronounced in spontaneous than medically indicated PTB. PTB alone in the absence of CA only had a moderate association with ASD and ADHD. Placental maternal vascular malperfusion alone or in combination with PTB was not associated with the risk of NDDs. DISCUSSION Our study provided new insights on PTB and NDDs by further considering preterm subtypes and placental histology. We revealed that children of spontaneous PTB along with histological CA were at the highest risk for a spectrum of NDDs.
Collapse
|
21
|
Bodnar TS, Raineki C, Wertelecki W, Yevtushok L, Plotka L, Zymak-Zakutnya N, Honerkamp-Smith G, Wells A, Rolland M, Woodward TS, Coles CD, Kable JA, Chambers CD, Weinberg J. Altered maternal immune networks are associated with adverse child neurodevelopment: Impact of alcohol consumption during pregnancy. Brain Behav Immun 2018; 73:205-215. [PMID: 29738852 PMCID: PMC6344127 DOI: 10.1016/j.bbi.2018.05.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 01/10/2023] Open
Abstract
Cytokines and chemokines are potent modulators of brain development and as such, dysregulation of the maternal immune system can result in deviations in the fetal cytokine balance, altering the course of typical brain development, and putting the individual on a "pathway to pathology". In the current study, we used a multi-variate approach to evaluate networks of interacting cytokines and investigated whether alterations in the maternal immune milieu could be linked to alcohol-related and alcohol-independent child neurodevelopmental delay. This was achieved through the measurement of 40 cytokines/chemokines from maternal blood samples collected during the second and third trimesters of pregnancy. Importantly, during the second trimester we identified network enrichment in levels of cytokines including IFN-ɣ, IL-10, TNF-β, TNF-α, and CRP associated with offspring neurodevelopmental delay. However, as elevations in levels of these cytokines have previously been reported in a wide range of neurodevelopmental disorders including autism spectrum disorder and schizophrenia, we suggest that this cytokine profile is likely not disorder specific, but rather may be an indicator of neurodevelopmental delay in general. By contrast, distinct clusters of activated/inhibited cytokines were identified based on maternal alcohol consumption and child neurodevelopmental outcome. Specifically, cytokines including IL-15, IL-10, MDC, and members of the VEGF sub-family were highest in alcohol-consuming mothers of children with neurodevelopmental delay and were identified in both network analyses and examination of individual cytokines, whereas a differential and unique cytokine profile was identified in the case of alcohol-independent child neurodevelopmental delay. We propose that the current findings could provide a critical step towards the development of early biomarkers and possibly interventions for alcohol-related neurodevelopmental delay. Importantly, the current approach could be informative for understanding mechanisms linking maternal immune system dysfunction and adverse child outcomes in a range of other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tamara S. Bodnar
- Department of Cellular and Physiological Sciences,
University of British Columbia, Vancouver, BC, Canada,Corresponding author: Tamara S.
Bodnar, Ph.D., Department of Cellular and Physiological Sciences, University of
British Columbia, 3307 – 2350 Health Sciences Mall, Vancouver, BC V6T
1Z3, Canada, , Phone: +1 (604) 822-4554, FAX:
+1 (604) 822-2316
| | - Charlis Raineki
- Department of Cellular and Physiological Sciences,
University of British Columbia, Vancouver, BC, Canada
| | | | - Lyubov Yevtushok
- OMNI-Net for Children International Charitable Fund, Rivne
Oblast Medical Diagnostic Center, Rivne, Ukraine
| | - Larisa Plotka
- OMNI-Net for Children International Charitable Fund, Rivne
Oblast Medical Diagnostic Center, Rivne, Ukraine
| | - Natalya Zymak-Zakutnya
- OMNI-Net for Children International Charitable Fund,
Khmelnytsky Perinatal Center, Khmelnytsky, Ukraine
| | | | - Alan Wells
- Department of Pediatrics, University of California San
Diego, La Jolla, USA
| | - Matthieu Rolland
- Department of Pediatrics, University of California San
Diego, La Jolla, USA
| | - Todd S. Woodward
- Department of Psychiatry, University of British Columbia,
Vancouver, Canada,Translational Research Unit, BC Mental Health and
Addictions Research Institute, Provincial Health Services Authority, Vancouver, BC,
Canada
| | - Claire D. Coles
- Department of Psychiatry and Behavioral Sciences;
Department of Pediatrics, Emory University School of Medicine, Atlanta, USA
| | - Julie A. Kable
- Department of Psychiatry and Behavioral Sciences;
Department of Pediatrics, Emory University School of Medicine, Atlanta, USA
| | - Christina D. Chambers
- Department of Pediatrics, University of California San
Diego, La Jolla, USA,Department of Family Medicine and Public Health, University
of California San Diego, La Jolla, CA, USA
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences,
University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
22
|
Kong L, Norstedt G, Schalling M, Gissler M, Lavebratt C. The Risk of Offspring Psychiatric Disorders in the Setting of Maternal Obesity and Diabetes. Pediatrics 2018; 142:peds.2018-0776. [PMID: 30093539 DOI: 10.1542/peds.2018-0776] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Prenatal exposure to metabolic disturbances is associated with increased risk of offspring neurodevelopmental impairment and autism spectrum disorder, while little is known about the joint effect of maternal obesity and diabetes. With this study, we aim to assess the joint effect of maternal obesity and diabetes on the risk for offspring psychiatric and mild neurodevelopmental disorders. METHODS Nationwide registries were used to link data of all live births in Finland between 2004 and 2014 (n = 649 043). Cox proportional hazards modeling adjusting for potential confounders was applied to estimate the effect of maternal obesity, pregestational diabetes mellitus (PGDM), and gestational diabetes mellitus, as well as their joint effects, on the outcomes of offspring psychiatric and mild neurodevelopmental diagnoses and offspring prescription of psychotropic drugs. RESULTS Among mothers without diabetes, severely obese mothers had 67% to 88% increased risk of having a child with mild neurodevelopmental disorders (hazard risk ratio [HR] = 1.69; 95% confidence interval [CI] = 1.54-1.86), attention-deficit/hyperactivity disorder or conduct disorder (HR = 1.88; 95% CI = 1.58-2.23), and psychotic, mood, and stress-related disorders (HR = 1.67; 95% CI = 1.31-2.13) compared with mothers with a normal BMI. PGDM implied a further risk increase for all groups of psychiatric diagnoses with onset in childhood or adolescence in mothers with severe obesity. Marked effects were found particularly for autism spectrum disorder (HR = 6.49; 95% CI = 3.08-13.69), attention-deficit/hyperactivity disorder and conduct disorder (HR = 6.03; 95% CI = 3.23-11.24), and mixed disorders of conduct and emotions (HR = 4.29; 95% CI = 2.14-8.60). Gestational diabetes mellitus did not increase the risk highly for these offspring disorders. CONCLUSIONS Maternal PGDM combined with severe maternal obesity markedly increases the risk of several children's psychiatric and mild neurodevelopmental disorders.
Collapse
Affiliation(s)
- Linghua Kong
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Gunnar Norstedt
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.,Department of Biochemistry, Sultan Qaboos University, Muscat, Oman
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mika Gissler
- Division of Family Medicine, Department of Neurobiology, Care Sciences, and Society and.,National Institute for Health and Welfare, Helsinki and Oulu, Finland; and.,Research Centre for Child Psychiatry, University of Turku, Turku, Finland
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; .,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
OH KJ, PARK JY, LEE J, HONG JS, ROMERO R, YOON BH. The combined exposure to intra-amniotic inflammation and neonatal respiratory distress syndrome increases the risk of intraventricular hemorrhage in preterm neonates. J Perinat Med 2018; 46:9-20. [PMID: 28672753 PMCID: PMC5848500 DOI: 10.1515/jpm-2016-0348] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/12/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To evaluate the impact of combined exposure to intra-amniotic inflammation and neonatal respiratory distress syndrome (RDS) on the development of intraventricular hemorrhage (IVH) in preterm neonates. METHODS This retrospective cohort study includes 207 consecutive preterm births (24.0-33.0 weeks of gestation). Intra-amniotic inflammation was defined as an amniotic fluid matrix metalloproteinase-8 concentration >23 ng/mL. According to McMenamin's classification, IVH was defined as grade II or higher when detected by neurosonography within the first weeks of life. RESULTS (1) IVH was diagnosed in 6.8% (14/207) of neonates in the study population; (2) IVH was frequent among newborns exposed to intra-amniotic inflammation when followed by postnatal RDS [33% (6/18)]. The frequency of IVH was 7% (8/115) among neonates exposed to either of these conditions - intra-amniotic inflammation or RDS - and 0% (0/64) among those who were not exposed to these conditions; and (3) Neonates exposed to intra-amniotic inflammation and postnatal RDS had a significantly higher risk of IVH than those with only intra-amniotic inflammation [odds ratio (OR) 4.6, 95% confidence interval (CI) 1.1-19.3] and those with RDS alone (OR 5.6, 95% CI 1.0-30.9), after adjusting for gestational age. CONCLUSION The combined exposure to intra-amniotic inflammation and postnatal RDS markedly increased the risk of IVH in preterm neonates.
Collapse
Affiliation(s)
- Kyung Joon OH
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea,Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam-si, Korea
| | - Jee Yoon PARK
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - JoonHo LEE
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Korea
| | - Joon-Seok HONG
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea,Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam-si, Korea
| | - Roberto ROMERO
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, USA, and Detroit, Michigan, USA,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Bo Hyun YOON
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
24
|
Coo H, Brownell MD, Ruth C, Flavin M, Au W, Day AG. Interpregnancy Interval and Congenital Anomalies: A Record-Linkage Study Using the Manitoba Population Research Data Repository. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2017; 39:996-1007. [PMID: 28757407 DOI: 10.1016/j.jogc.2017.04.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/28/2017] [Accepted: 04/26/2017] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Congenital anomalies are a serious public health issue, and relatively few modifiable risk factors have been identified. Our objective was to investigate one such potential risk factor, the interpregnancy interval (IPI). METHODS We conducted a secondary analysis of data housed at the Manitoba Centre for Health Policy. In-hospital live births and stillbirths of at least 20 weeks' gestation were identified, and consecutive births to the same mother were grouped into sibling pairs to calculate the IPI for the younger siblings of each pair. Logistic regression models were fit to examine the association between the IPI and any congenital anomaly, as well as CNS and chromosomal anomalies, while controlling for potentially confounding sociodemographic and clinical factors. RESULTS Among 172 909 live births and stillbirths, the IPI was not significantly associated with congenital anomalies overall or with chromosomal anomalies. Short IPIs were associated with significantly increased odds of CNS anomalies relative to IPIs of 18-23 months (adjusted OR [aOR] for IPIs <6 months 2.15; 95% CI 1.48-3.12), whereas the aOR for IPIs ≥60 months was elevated but not statistically significant (aOR 1.50; 95% CI 0.96-2.34). In a sensitivity analysis in which the cohort was restricted to births from 2003 onwards (which yielded more complete data on health-related behaviours), the observed effect for IPIs shorter than 6 months and CNS anomalies was attenuated and no longer significant, but it remained elevated (aOR 1.65; 95% CI 0.85-3.24). CONCLUSION The findings for CNS anomalies warrant further investigation.
Collapse
Affiliation(s)
- Helen Coo
- Department of Pediatrics, Queen's University, Kingston, ON.
| | - Marni D Brownell
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB; Manitoba Centre for Health Policy, Winnipeg, MB
| | - Chelsea Ruth
- Manitoba Centre for Health Policy, Winnipeg, MB; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB
| | - Michael Flavin
- Department of Pediatrics, Queen's University, Kingston, ON
| | - Wendy Au
- Manitoba Centre for Health Policy, Winnipeg, MB
| | - Andrew G Day
- Kingston General Hospital Research Institute, Kingston, ON
| |
Collapse
|
25
|
Qanitha A, de Mol BAJM, Burgner DP, Kabo P, Pabittei DR, Yusuf I, Uiterwaal CSPM. Pregnancy-related conditions and premature coronary heart disease in adult offspring. HEART ASIA 2017; 9:90-95. [PMID: 29259659 PMCID: PMC5730950 DOI: 10.1136/heartasia-2017-010896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 12/29/2022]
Abstract
Objective To investigate the association between complications during pregnancy and premature coronary heart disease in adult offspring. Methods We conducted a population-based case-control study of 153 Indonesian patients with a first acute coronary syndrome (ACS) (age ≤55 years) and 153 age-matched and sex-matched controls. Data on complications during pregnancy (high blood pressure, preterm delivery) and maternal infections in pregnancy were obtained, together with sociodemographic data, clinical profiles, laboratory measurements and adulthood cardiovascular disease (CVD) risk factors at hospital admission or enrolment. Conditional logistic regression was performed to assess the association between overall pregnancy complications, and specific groupings of complications and premature ACS. Results Pregnancy-related hypertension and infection were more common in mothers of cases than controls. Pregnancy complications were associated with premature offspring ACS (OR 2.9, 95% CI 1.4 to 6.0, p=0.004), and the association persisted in fully adjusted analyses (ORadjusted 4.5, 1.1 to 18.1, p=0.036). In subgroup analyses, pregnancy-related high blood pressure (ORadjusted 5.0, 1.0 to 24.7, p=0.050) and maternal infections (ORadjusted 5.2, 1.1 to 24.2, p=0.035) were associated with offspring ACS. Conclusions Offspring of mothers with complications during pregnancy have an increased risk for premature ACS in adulthood, which may be of particular relevance in populations in transition, where the incidence of both pregnancy-related morbidity and CVD are high.
Collapse
Affiliation(s)
- Andriany Qanitha
- Department of Cardio-thoracic Surgery, AMC Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Physiology, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Bastianus A J M de Mol
- Department of Cardio-thoracic Surgery, AMC Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - David P Burgner
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Peter Kabo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Dara R Pabittei
- Department of Cardio-thoracic Surgery, AMC Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Physiology, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Irawan Yusuf
- Department of Physiology, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Cuno S P M Uiterwaal
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
26
|
Ginsberg Y, Khatib N, Weiner Z, Beloosesky R. Maternal Inflammation, Fetal Brain Implications and Suggested Neuroprotection: A Summary of 10 Years of Research in Animal Models. Rambam Maimonides Med J 2017; 8:RMMJ.10305. [PMID: 28467756 PMCID: PMC5415374 DOI: 10.5041/rmmj.10305] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A growing body of evidence implies that maternal inflammation during pregnancy is associated with increased risk of neurodevelopmental disorders in the offspring. The pathophysiological mechanisms by which maternal inflammation evokes fetal brain injury and contributes to long-term adverse neurological outcomes are not completely understood. In this review, we summarize 10 years of our research experience on maternal inflammation and the implications upon the fetal/offspring brain. We review our findings regarding the underlying mechanisms that connects maternal inflammation and fetal brain injuries (e.g. cytokines, oxidative stress); we discuss our imaging, pathological and behavioral test results which support brain damage following maternal inflammation; and finally we describe some of the therapeutic strategies which might prevent the damage.
Collapse
Affiliation(s)
- Yuval Ginsberg
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
| | - Nizar Khatib
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
- The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ron Beloosesky
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
- The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
27
|
Gabriel ML, Braga FB, Cardoso MR, Lopes AC, Piatto VB, Souza AS. The association between pro- and anti-inflammatory cytokine polymorphisms and periventricular leukomalacia in newborns with hypoxic-ischemic encephalopathy. J Inflamm Res 2016; 9:59-67. [PMID: 27217792 PMCID: PMC4862342 DOI: 10.2147/jir.s103697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Periventricular leukomalacia (PVL) is a frequent consequence of hypoxic-ischemic injury. Functional cytokine gene variants that result in altered production of inflammatory (tumor necrosis factor-alpha [TNF-α] and interleukin-1beta [IL-1β]) or anti-inflammatory (interleukin-10 [IL-10]) cytokines may modify disease processes, including PVL. Objective The aim of this study was to evaluate if there is a relationship between the two proinflammatory polymorphisms (TNF-α-1031T/C and IL-1β-511C/T) and the anti-inflammatory polymorphism IL-10-1082G/A and PVL risk in Brazilian newborns with and without this injury. Materials and methods A cross-sectional case-control study performed at the Neonatal Intensive Care Unit of the Children’s Hospital and Maternity of the São José do Rio Preto Medical School (FAMERP). Fifty preterm and term newborns were examined as index cases and 50 term newborns as controls, of both sexes for both groups. DNA was extracted from peripheral blood leukocytes, and the sites that encompassed the three polymorphisms were amplified by polymerase chain reaction-restriction fragment length polymorphism. Results Gestational age ranged from 25 to 39 weeks, in the case group, and in the control group it ranged from 38 to 42.5 weeks (P<0.0001). Statistically significant association was found between TNF-α-1031T/C high expression genotype TC (odds ratio [OR], 2.495; 95% confidence interval [CI], 1.10–5.63; P=0.043) as well as between genotypes (TC + CC) (OR, 2.471; 95% CI, 1.10–5.55; P=0.044) and risk of PVL. Statistically significant association was found between IL-1β-511C/T high expression genotypes (CT + TT) (OR, 23.120; 95% CI, 1.31–409.4; P=0.003) and risk of PVL. Statistically significant association between IL-10-1082G/A high expression genotype GG (OR, 0.07407; 95% CI, 0.02–0.34; P<0.0001) as well as between IL-10-1082G high expression allele (OR, 0.5098; 95% CI, 0.29–0.91; P=0,030) and PVL reduced risk was observed. There was a statistically significant association between TC/CT/GA genotype combination and the risk of PVL (OR, 6.469; 95% CI, 2.00–20.92; P=0.001). Conclusion There is evidence of an association between the polymorphisms TNF-α-1031T/C, IL-1β-511C/T, and IL-10-1082G/A and PVL risk in this Brazilian newborn population studied.
Collapse
Affiliation(s)
- Marta Lúcia Gabriel
- Radiology Department, São José do Rio Preto Medical School, FAMERP, São Paulo, Brazil
| | | | | | - Ana Cláudia Lopes
- Morphology Department, São José do Rio Preto Medical School, FAMERP, São Paulo, Brazil
| | | | - Antônio Soares Souza
- Radiology Department, São José do Rio Preto Medical School, FAMERP, São Paulo, Brazil
| |
Collapse
|
28
|
Wang X, Wang J, Luo H, Chen C, Pei F, Cai Y, Yang X, Wang N, Fu J, Xu Z, Zhou L, Zeng C. Prenatal lipopolysaccharide exposure causes mesenteric vascular dysfunction through the nitric oxide and cyclic guanosine monophosphate pathway in offspring. Free Radic Biol Med 2015; 86:322-30. [PMID: 26073126 DOI: 10.1016/j.freeradbiomed.2015.05.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/03/2015] [Accepted: 05/26/2015] [Indexed: 01/21/2023]
Abstract
Cardiovascular diseases, such as hypertension, could be programmed in fetal life. Prenatal lipopolysaccharide (LPS) exposure in utero results in increased blood pressure in offspring, but the vascular mechanisms involved are unclear. Pregnant Sprague-Dawley rats were intraperitoneally injected with LPS (0.79mg/kg) or saline (0.5ml) on gestation days 8, 10, and 12. The offspring of LPS-treated dams had higher blood pressure and decreased acetylcholine (ACh)-induced relaxation and increased phenylephrine (PE)-induced contraction in endothelium-intact mesenteric arteries. Endothelium removal significantly enhanced the PE-induced contraction in offspring of control but not LPS-treated dams. The arteries pretreated with l-NAME to inhibit nitric oxide synthase (eNOS) in the endothelium or ODQ to inhibit cGMP production in the vascular smooth muscle had attenuated ACh-induced relaxation but augmented PE-induced contraction to a larger extent in arteries from offspring of control than those from LPS-treated dams. In addition, the endothelium-independent relaxation caused by sodium nitroprusside was also decreased in arteries from offspring of LPS-treated dams. The functional results were accompanied by a reduction in the expressions of eNOS and soluble guanylate cyclase (sGC) and production of NO and cGMP in arteries from offspring of LPS-treated dams. Furthermore, LPS-treated dam's offspring arteries had increased oxidative stress and decreased antioxidant capacity. Three-week treatment with TEMPOL, a reactive oxygen species (ROS) scavenger, normalized the alterations in the levels of ROS, eNOS, and sGC, as well as in the production of NO and cGMP and vascular function in the arteries of the offspring of LPS-treated dams. In conclusion, prenatal LPS exposure programs vascular dysfunction of mesenteric arteries through increased oxidative stress and impaired NO-cGMP signaling pathway.
Collapse
Affiliation(s)
- Xinquan Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Fang Pei
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Yue Cai
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Xiaoli Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Na Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Jinjuan Fu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Zaichen Xu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Lin Zhou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China.
| |
Collapse
|
29
|
Intra-Amniotic LPS Induced Region-Specific Changes in Presynaptic Bouton Densities in the Ovine Fetal Brain. BIOMED RESEARCH INTERNATIONAL 2015; 2015:276029. [PMID: 26417592 PMCID: PMC4568354 DOI: 10.1155/2015/276029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 12/14/2014] [Indexed: 11/17/2022]
Abstract
Rationale. Chorioamnionitis has been associated with increased risk for fetal brain damage. Although, it is now accepted that synaptic dysfunction might be responsible for functional deficits, synaptic densities/numbers after a fetal inflammatory challenge have not been studied in different regions yet. Therefore, we tested in this study the hypothesis that LPS-induced chorioamnionitis caused profound changes in synaptic densities in different regions of the fetal sheep brain. Material and Methods. Chorioamnionitis was induced by a 10 mg intra-amniotic LPS injection at two different exposure intervals. The fetal brain was studied at 125 days of gestation (term = 150 days) either 2 (LPS2D group) or 14 days (LPS14D group) after LPS or saline injection (control group). Synaptophysin immunohistochemistry was used to quantify the presynaptic density in layers 2-3 and 5-6 of the motor cortex, somatosensory cortex, entorhinal cortex, and piriforme cortex, in the nucleus caudatus and putamen and in CA1/2, CA3, and dentate gyrus of the hippocampus. Results. There was a significant reduction in presynaptic bouton densities in layers 2-3 and 5-6 of the motor cortex and in layers 2-3 of the entorhinal and the somatosensory cortex, in the nucleus caudate and putamen and the CA1/2 and CA3 of the hippocampus in the LPS2D compared to control animals. Only in the motor cortex and putamen, the presynaptic density was significantly decreased in the LPS14 D compared to the control group. No changes were found in the dentate gyrus of the hippocampus and the piriforme cortex. Conclusion. We demonstrated that LPS-induced chorioamnionitis caused a decreased density in presynaptic boutons in different areas in the fetal brain. These synaptic changes seemed to be region-specific, with some regions being more affected than others, and seemed to be transient in some regions.
Collapse
|
30
|
Jo H, Schieve LA, Sharma AJ, Hinkle SN, Li R, Lind JN. Maternal prepregnancy body mass index and child psychosocial development at 6 years of age. Pediatrics 2015; 135:e1198-209. [PMID: 25917989 PMCID: PMC4411780 DOI: 10.1542/peds.2014-3058] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2015] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Both obesity and developmental disabilities have increased in recent decades. Limited studies suggest associations between maternal prepregnancy obesity and child neurodevelopment. METHODS The Infant Feeding Practices Study II, a US nationally distributed longitudinal study of maternal health and infant health and feeding practices, was conducted from 2005 to 2007. In 2012, mothers were recontacted for information on their children's health and development. We examined associations between maternal prepregnancy BMI and child psychosocial development in 1311 mother-child pairs included in this follow-up study. Children's development was assessed by maternal report of child psychosocial difficulties from the Strengths and Difficulties Questionnaire, past developmental diagnoses, and receipt of special needs services. RESULTS Adjusting for sociodemographic factors, children of obese class II/III mothers (BMI >35.0) had increased odds of emotional symptoms (adjusted odds ratio [aOR] 2.24; 95% confidence interval [CI], 1.27-3.98), peer problems (aOR 2.07; 95% CI, 1.26-3.40), total psychosocial difficulties (aOR 2.17; 95% CI, 1.24-3.77), attention-deficit/hyperactivity disorder diagnosis (aOR 4.55; 95% CI, 1.80-11.46), autism or developmental delay diagnosis (aOR 3.13; 95% CI, 1.10-8.94), receipt of speech language therapy (aOR 1.93; 95% CI, 1.18-3.15), receipt of psychological services (aOR 2.27; 95% CI, 1.09-4.73), and receipt of any special needs service (aOR 1.99; 95% CI, 1.33-2.97) compared with children of normal weight mothers (BMI 18.5-24.9). Adjustment for potential causal pathway factors including pregnancy weight gain, gestational diabetes, breastfeeding duration, postpartum depression, and child's birth weight did not substantially affect most estimates. CONCLUSIONS Children whose mothers were severely obese before pregnancy had increased risk for adverse developmental outcomes.
Collapse
Affiliation(s)
- Heejoo Jo
- Division of Birth Defects and Developmental Disabilities, National Center on Birth Defects and Developmental Disabilities, Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee;
| | - Laura A Schieve
- Division of Birth Defects and Developmental Disabilities, National Center on Birth Defects and Developmental Disabilities
| | - Andrea J Sharma
- Division of Reproductive Health, National Center for Chronic Disease Prevention and Health Promotion, Atlanta, Georgia; United States Public Health Service Commissioned Corps, Atlanta, Georgia; and
| | - Stefanie N Hinkle
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Ruowei Li
- Division of Nutrition, Physical Activity, and Obesity, and
| | - Jennifer N Lind
- United States Public Health Service Commissioned Corps, Atlanta, Georgia; and Division of Nutrition, Physical Activity, and Obesity, and Epidemic Intelligence Service, Office of Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
31
|
Rocha-Ferreira E, Hristova M. Antimicrobial peptides and complement in neonatal hypoxia-ischemia induced brain damage. Front Immunol 2015; 6:56. [PMID: 25729383 PMCID: PMC4325932 DOI: 10.3389/fimmu.2015.00056] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/29/2015] [Indexed: 12/22/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a clinical condition in the neonate, resulting from oxygen deprivation around the time of birth. HIE affects 1-5/1000 live births worldwide and is associated with the development of neurological deficits, including cerebral palsy, epilepsy, and cognitive disabilities. Even though the brain is considered as an immune-privileged site, it has innate and adaptive immune response and can produce complement (C) components and antimicrobial peptides (AMPs). Dysregulation of cerebral expression of AMPs and C can exacerbate or ameliorate the inflammatory response within the brain. Brain ischemia triggers a prolonged inflammatory response affecting the progression of injury and secondary energy failure and involves both innate and adaptive immune systems, including immune-competent and non-competent cells. Following injury to the central nervous system (CNS), including neonatal hypoxia-ischemia (HI), resident microglia, and astroglia are the main cells providing immune defense to the brain in a stimulus-dependent manner. They can express and secrete pro-inflammatory cytokines and therefore trigger prolonged inflammation, resulting in neurodegeneration. Microglial cells express and release a wide range of inflammation-associated molecules including several components of the complement system. Complement activation following neonatal HI injury has been reported to contribute to neurodegeneration. Astrocytes can significantly affect the immune response of the CNS under pathological conditions through production and release of pro-inflammatory cytokines and immunomodulatory AMPs. Astrocytes express β-defensins, which can chemoattract and promote maturation of dendritic cells (DC), and can also limit inflammation by controlling the viability of these same DC. This review will focus on the balance of complement components and AMPs within the CNS following neonatal HI injury and the effect of that balance on the subsequent brain damage.
Collapse
Affiliation(s)
- Eridan Rocha-Ferreira
- Perinatal Brain Repair Group, Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , UK
| | - Mariya Hristova
- Perinatal Brain Repair Group, Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London , London , UK
| |
Collapse
|
32
|
Wang X, Luo H, Chen C, Chen K, Wang J, Cai Y, Zheng S, Yang X, Zhou L, Jose PA, Zeng C. Prenatal lipopolysaccharide exposure results in dysfunction of the renal dopamine D1 receptor in offspring. Free Radic Biol Med 2014; 76:242-50. [PMID: 25236748 PMCID: PMC6873924 DOI: 10.1016/j.freeradbiomed.2014.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/23/2014] [Accepted: 08/11/2014] [Indexed: 12/31/2022]
Abstract
Adverse environment in early life can modulate the adult phenotype, including blood pressure. Lipopolysaccharide (LPS) exposure in utero results in increased blood pressure in the offspring, but the exact mechanisms are not clear. Studies have shown that the renal dopamine D1 receptor (D1R) plays an important role in maintaining sodium homeostasis and normal blood pressure; dysfunction of D1R is associated with oxidative stress and hypertension. In this study, we determined if dysfunction of the renal D1R is involved in fetal-programmed hypertension, and if oxidative stress contributes to this process. Pregnant Sprague-Dawley (SD) rats were intraperitoneally injected with LPS (0.79 mg/kg) or saline at gestation days 8, 10, and 12. As compared with saline-injected (control) dams, offspring of LPS-treated dams had increased blood pressure, decreased renal sodium excretion, and increased markers of oxidative stress. In addition, offspring of LPS-treated dams had decreased renal D1R expression, increased D1R phosphorylation, and G protein-coupled receptor kinase type 2 (GRK2) and type 4 (GRK4) protein expression, and impaired D1R-mediated natriuresis and diuresis. All of the findings in the offspring of LPS-treated dams were normalized after treatment with TEMPOL, an oxygen free radical scavenger. In conclusion, prenatal LPS exposure, via an increase in oxidative stress, impairs renal D1R function and leads to hypertension in the offspring. Normalization of renal D1R function by amelioration of oxidative stress may be a therapeutic target of fetal programming of hypertension.
Collapse
Affiliation(s)
- Xinquan Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Ken Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Yue Cai
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Xiaoli Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Lin Zhou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China.
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, People's Republic of China; Chongqing Institute of Cardiology, Chongqing, People's Republic of China.
| |
Collapse
|
33
|
Huang L, Yu X, Keim S, Li L, Zhang L, Zhang J. Maternal prepregnancy obesity and child neurodevelopment in the Collaborative Perinatal Project. Int J Epidemiol 2014; 43:783-92. [PMID: 24569381 DOI: 10.1093/ije/dyu030] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To examine the association between maternal prepregnancy weight and child neurodevelopment, and the effect of gestational weight gain. METHODS Using the U.S. Collaborative Perinatal Project data, 1959-76, a total of 30,212 women with a calculable prepregnancy body mass index (BMI) and gestational weight gain, and term singleton children followed up for more than 7 years were included in this study. Intelligence quotient (IQ) was measured at 7 years of age by Wechsler Intelligence Scales. RESULTS Maternal prepregnancy BMI displayed inverted U-shaped associations with child IQ after adjustment for maternal age, maternal education levels, maternal race, marital status, socioeconomic status, smoking during pregnancy, parity and study center. Women with BMI at around 20 kg/m2 appeared to have the highest offspring IQ scores. After controlling for familial factors in the siblings' sample, maternal obesity (BMI≥30.0 kg/m2) was associated with lower Full-scale IQ (adjusted ß=-2.0, 95% confidence interval -3.5 to -0.5), and Verbal scale IQ (adjusted ß=-2.5, 95% confidence interval -4.0 to -1.0), using BMI of 18.5-24.9 kg/m2 as the reference category. Compared with children born to normal-weight women who gained 21-25 lb. during pregnancy, those born to obese women who gained more than 40 lb. had 6.5 points deficit in IQ after adjustment for potential confounders. CONCLUSIONS Maternal prepregnancy obesity was associated with lower child IQ, and excessive weight gain accelerated the association. With obesity rising steadily, these results appear to raise serious public health concerns.
Collapse
Affiliation(s)
- Lisu Huang
- MOE-Shanghai Key Laboratory of Children's Environmental Health and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Research Institute at Nationwide Children's Hospital, Center for Biobehavioral Health, Columbus, OH, USA and Department of Pediatrics and Division of Epidemiology, Ohio State University, Columbus, OH, USA and Department of Obstetrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, ChinaMOE-Shanghai Key Laboratory of Children's Environmental Health and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Research Institute at Nationwide Children's Hospital, Center for Biobehavioral Health, Columbus, OH, USA and Department of Pediatrics and Division of Epidemiology, Ohio State University, Columbus, OH, USA and Department of Obstetrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodan Yu
- MOE-Shanghai Key Laboratory of Children's Environmental Health and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Research Institute at Nationwide Children's Hospital, Center for Biobehavioral Health, Columbus, OH, USA and Department of Pediatrics and Division of Epidemiology, Ohio State University, Columbus, OH, USA and Department of Obstetrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sarah Keim
- MOE-Shanghai Key Laboratory of Children's Environmental Health and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Research Institute at Nationwide Children's Hospital, Center for Biobehavioral Health, Columbus, OH, USA and Department of Pediatrics and Division of Epidemiology, Ohio State University, Columbus, OH, USA and Department of Obstetrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Li
- MOE-Shanghai Key Laboratory of Children's Environmental Health and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Research Institute at Nationwide Children's Hospital, Center for Biobehavioral Health, Columbus, OH, USA and Department of Pediatrics and Division of Epidemiology, Ohio State University, Columbus, OH, USA and Department of Obstetrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Research Institute at Nationwide Children's Hospital, Center for Biobehavioral Health, Columbus, OH, USA and Department of Pediatrics and Division of Epidemiology, Ohio State University, Columbus, OH, USA and Department of Obstetrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Research Institute at Nationwide Children's Hospital, Center for Biobehavioral Health, Columbus, OH, USA and Department of Pediatrics and Division of Epidemiology, Ohio State University, Columbus, OH, USA and Department of Obstetrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Mann JR, Deroche CB, Spiryda LB, McDermott S. Is pre-pregnancy weight a risk factor for intellectual disability? ACTA ACUST UNITED AC 2014. [DOI: 10.1586/eog.13.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Barateiro A, Domingues HS, Fernandes A, Relvas JB, Brites D. Rat Cerebellar Slice Cultures Exposed to Bilirubin Evidence Reactive Gliosis, Excitotoxicity and Impaired Myelinogenesis that Is Prevented by AMPA and TNF-α Inhibitors. Mol Neurobiol 2013; 49:424-39. [DOI: 10.1007/s12035-013-8530-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/05/2013] [Indexed: 12/11/2022]
|
36
|
Tian X, Zhu M, Du L, Wang J, Fan Z, Liu J, Zhao Y, Nie G. Intrauterine inflammation increases materno-fetal transfer of gold nanoparticles in a size-dependent manner in murine pregnancy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:2432-2439. [PMID: 23761193 DOI: 10.1002/smll.201300817] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/24/2013] [Indexed: 06/02/2023]
Abstract
The materno-fetal transfer of nanoparticles is a critical issue in designing theranoustic nanoparticles for in vivo applications during pregnancy. Recent studies have reported that certain nanoparticles can cross the placental barrier in healthy pregnant animals depending on the size and surface modification of the nanoparticles and the developmental stages of the fetuses. However, materno-fetal transfer under pathological pregnant conditions has not been examined so far. Here, it is shown that intrauterine inflammation can enhance the materno-fetal transfer of nanoparticles in the late gestation stage of murine pregnancy in a size-dependent manner. Three different-sized gold nanoparticles (Au NPs) with diameters of 3 (Au3), 13 (Au13) and 32 (Au32) nm are applied. The accumulation of Au3 and Au13 nanoparticles in the fetuses is significantly increased in intrauterine inflammatory mice compared with healthy control mice: the concentration of Au3 is much higher than Au13 in fetal tissues of intrauterine inflammatory mice. In contrast, Au32 nanoparticles cannot cross the placental barrier either in healthy or in intrauterine inflammatory mice. The possible underlying mechanism of the increased materno-fetal transfer of small-sized nanoparticles on pathological conditions is inferred to be the structural and functional abnormalities of the placenta under intrauterine inflammation. The size of the nanoparticles is one of the critical factors which determines the extent of fetal exposure to nanoparticles in murine pregnancy under inflammatory conditions.
Collapse
Affiliation(s)
- Xin Tian
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China Beijing 100190, China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Maternal prepregnancy weight status and associations with children's development and disabilities at kindergarten. Int J Obes (Lond) 2013; 37:1344-51. [PMID: 23860335 DOI: 10.1038/ijo.2013.128] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 07/02/2013] [Accepted: 07/04/2013] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Obesity is prevalent among women of reproductive age, and developmental disabilities in children continue to increase. We examined associations between mother's prepregnancy body mass index (BMI) and physical and developmental disabilities, and objective measures of reading and math skills and fine and gross motor function in children. METHODS We used the Early Childhood Longitudinal Study-Birth Cohort (ECLS-B; n=5200), a cohort of children born in 2001 and followed until kindergarten. Children were classified according to maternal prepregnancy BMI (in kg per m(2)): underweight (BMI <18.5), normal weight (BMI 18.5-24.9), overweight (BMI 25.0-29.9), obese class I (BMI 30.0-34.9) and obese class II/III (BMI ≥ 35.0). Parent reports of doctor-diagnosed disabilities were collected up to kindergarten and classified as learning and behavioral or physical. Children's reading and math and fine and gross motor function were assessed at kindergarten according to standardized tests. Linear and modified logistic regression models were adjusted for maternal sociodemographic variables, family enrichment variables, and children's sex, age and year of kindergarten entry. Additional adjustment for current child BMI was performed in separate models. All data are weighted to be nationally representative of the children born in 2001. RESULTS Compared with children of normal-weight mothers, children born to obese class II/III mothers had an increased risk of learning or behavioral (risk ratio 1.67; 95% confidence interval 1.27, 2.21)), but not physical disabilities (risk ratio 0.57; 95% confidence interval 0.27, 1.22). Gross (P<0.001), but not fine (P=0.06) motor function was significantly associated with maternal BMI, but gross motor function was attenuated after adjustment for current child BMI (P=0.05). Children's reading scores (P=0.01) but not math scores (P=0.11) were significantly associated with maternal BMI. CONCLUSIONS In this nationally representative US cohort, children born to severely obese mothers had an increased risk for diagnosed learning and behavioral but not physical disabilities by kindergarten.
Collapse
|
38
|
Rogers LK, Valentine CJ, Keim SA. DHA supplementation: current implications in pregnancy and childhood. Pharmacol Res 2013; 70:13-9. [PMID: 23266567 PMCID: PMC3602397 DOI: 10.1016/j.phrs.2012.12.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 11/07/2012] [Accepted: 12/10/2012] [Indexed: 02/08/2023]
Abstract
Dietary supplementation with ω-3 long chain fatty acids including docosahexaenoic acid (DHA) has increased in popularity in recent years and adequate DHA supplementation during pregnancy and early childhood is of clinical importance. Some evidence has been built for the neuro-cognitive benefits of supplementation with long chain polyunsaturated fatty acids (LCPUFA) such as DHA during pregnancy; however, recent data indicate that the anti-inflammatory properties may be of at least equal significance. Adequate DHA availability in the fetus/infant optimizes brain and retinal maturation in part by influencing neurotransmitter pathways. The anti-inflammatory properties of LCPUFA are largely mediated through modulation of signaling either directly through binding to receptors or through changes in lipid raft formation and receptor presentation. Our goal is to review the current findings on DHA supplementation, specifically in pregnancy and infant neurodevelopment, as a pharmacologic agent with both preventative and therapeutic value. Given the overall benefits of DHA, maternal and infant supplementation may improve neurological outcomes especially in vulernable populations. However, optimal composition of the supplement and dosing and treatment strategies still need to be determined to lend support for routine supplementation.
Collapse
Affiliation(s)
- Lynette K Rogers
- Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Dr., Department of Pediatrics, The Ohio State University, Columbus, OH, United States.
| | | | | |
Collapse
|
39
|
Kosmac K, Bantug GR, Pugel EP, Cekinovic D, Jonjic S, Britt WJ. Glucocorticoid treatment of MCMV infected newborn mice attenuates CNS inflammation and limits deficits in cerebellar development. PLoS Pathog 2013; 9:e1003200. [PMID: 23505367 PMCID: PMC3591306 DOI: 10.1371/journal.ppat.1003200] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 01/08/2013] [Indexed: 01/07/2023] Open
Abstract
Infection of the developing fetus with human cytomegalovirus (HCMV) is a major cause of central nervous system disease in infants and children; however, mechanism(s) of disease associated with this intrauterine infection remain poorly understood. Utilizing a mouse model of HCMV infection of the developing CNS, we have shown that peripheral inoculation of newborn mice with murine CMV (MCMV) results in CNS infection and developmental abnormalities that recapitulate key features of the human infection. In this model, animals exhibit decreased granule neuron precursor cell (GNPC) proliferation and altered morphogenesis of the cerebellar cortex. Deficits in cerebellar cortical development are symmetric and global even though infection of the CNS results in a non-necrotizing encephalitis characterized by widely scattered foci of virus-infected cells with mononuclear cell infiltrates. These findings suggested that inflammation induced by MCMV infection could underlie deficits in CNS development. We investigated the contribution of host inflammatory responses to abnormal cerebellar development by modulating inflammatory responses in infected mice with glucocorticoids. Treatment of infected animals with glucocorticoids decreased activation of CNS mononuclear cells and expression of inflammatory cytokines (TNF-α, IFN-β and IFNγ) in the CNS while minimally impacting CNS virus replication. Glucocorticoid treatment also limited morphogenic abnormalities and normalized the expression of developmentally regulated genes within the cerebellum. Importantly, GNPC proliferation deficits were normalized in MCMV infected mice following glucocorticoid treatment. Our findings argue that host inflammatory responses to MCMV infection contribute to deficits in CNS development in MCMV infected mice and suggest that similar mechanisms of disease could be responsible for the abnormal CNS development in human infants infected in-utero with HCMV.
Collapse
Affiliation(s)
- Kate Kosmac
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America.
| | | | | | | | | | | |
Collapse
|
40
|
Maternal conditions and perinatal characteristics associated with autism spectrum disorder and intellectual disability. PLoS One 2013; 8:e50963. [PMID: 23308096 PMCID: PMC3538698 DOI: 10.1371/journal.pone.0050963] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 10/29/2012] [Indexed: 01/29/2023] Open
Abstract
Background As well as being highly comorbid conditions, autism spectrum disorders (ASD) and intellectual disability (ID) share a number of clinically-relevant phenomena. This raises questions about similarities and overlap in diagnosis and aetiological pathways that may exist for both conditions. Aims To examine maternal conditions and perinatal factors for children diagnosed with an ASD, with or without ID, and children with ID of unknown cause, compared with unaffected children. Methods The study population comprised all live singleton births in Western Australia (WA) between January 1984 and December 1999 (N = 383,153). Univariate and multivariate multinomial logistic regression models were applied using a blocked modelling approach to assess the effect of maternal conditions, sociodemographic factors, labour and delivery characteristics and neonatal outcomes. Results In univariate analyses mild-moderate ID was associated with pregnancy hypertension, asthma, urinary tract infection, some types of ante-partum haemorrhage, any type of preterm birth, elective C-sections, breech presentation, poor fetal growth and need for resuscitation at birth, with all factors showing an increased risk. Severe ID was positively associated with poor fetal growth and need for resuscitation, as well as any labour or delivery complication. In the multivariate analysis no maternal conditions or perinatal factors were associated with an increased risk of ASD without ID. However, pregnancy hypertension and small head circumference were associated with a reduced risk (OR = 0.64, 95% CI: 0.43, 0.94; OR = 0.58, 95% CI: 0.34, 0.96, respectively). For ASD with ID, threatened abortion before 20 weeks gestation and poor fetal growth were associated with an increased risk. Conclusion Findings show that indicators of a poor intrauterine environment are associated with an elevated risk of ID, while for ASD, and particularly ASD without ID, the associations are much weaker. As such, these findings highlight the importance of accounting for the absence or presence of ID when examining ASD, if we are to improve our understanding of the causal pathways associated with these conditions.
Collapse
|
41
|
Mann JR, McDermott SW, Hardin J, Pan C, Zhang Z. Pre-pregnancy body mass index, weight change during pregnancy, and risk of intellectual disability in children. BJOG 2012. [DOI: 10.1111/1471-0528.12052] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- JR Mann
- University of South Carolina School of Medicine; Columbia; SC; USA
| | - SW McDermott
- University of South Carolina School of Medicine; Columbia; SC; USA
| | - J Hardin
- University of South Carolina Arnold School of Public Health; Columbia; SC; USA
| | - C Pan
- University of South Carolina Arnold School of Public Health; Columbia; SC; USA
| | - Z Zhang
- University of South Carolina School of Medicine; Columbia; SC; USA
| |
Collapse
|
42
|
Hinkle SN, Schieve LA, Stein AD, Swan DW, Ramakrishnan U, Sharma AJ. Associations between maternal prepregnancy body mass index and child neurodevelopment at 2 years of age. Int J Obes (Lond) 2012; 36:1312-9. [PMID: 22964791 PMCID: PMC4583192 DOI: 10.1038/ijo.2012.143] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Both underweight and obese mothers have an increased risk for adverse offspring outcomes. Few studies have examined the association between prepregnancy body mass index (BMI) and children's neurodevelopment. SUBJECTS We used data from the nationally representative Early Childhood Longitudinal Study-Birth Cohort (ECLS-B; n=6850). Children were classified according to their mother's prepregnancy BMI (kg m(-2)) status: underweight (BMI <18.5), normal weight (BMI 18.5-24.9), overweight (BMI 25.0-29.9), obese class I (BMI 30.0-34.9), and obese class II and III (BMI ≥35.0). Children's age-adjusted mental development index (MDI) and psychomotor development index (PDI) T-scores (mean 50, s.d. 10) were obtained using a validated shortened version of the Bayley Scales of Infant Development-II at approximately 2 years of age. While adjusting for sociodemographics, we estimated the average MDI and PDI scores or the risk of delayed (<-1 s.d. vs >1 s.d.) mental or motor development, relative to children of normal weight mothers. RESULTS Compared with children of normal weight mothers, MDI scores were lower among children of mothers of all other prepregnancy BMI categories, with the greatest adjusted difference among children of class II and III obese mothers (-2.13 (95% CI -3.32, -0.93)). The adjusted risk of delayed mental development was increased among children of underweight (risk ratio (RR) 1.36 (95% CI 1.04, 1.78)) and class II and III obese (RR 1.38 (95% CI 1.03, 1.84)) mothers. Children's PDI scores or motor delay did not differ by maternal prepregnancy BMI. CONCLUSION In this nationally representative sample of 2-year-old US children, low and very-high maternal prepregnancy BMI were associated with increased risk of delayed mental development but not motor development.
Collapse
Affiliation(s)
- S N Hinkle
- Nutrition and Health Sciences, Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA
| | | | | | | | | | | |
Collapse
|
43
|
Rana SA, Aavani T, Pittman QJ. Sex effects on neurodevelopmental outcomes of innate immune activation during prenatal and neonatal life. Horm Behav 2012; 62:228-36. [PMID: 22516179 PMCID: PMC3522744 DOI: 10.1016/j.yhbeh.2012.03.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/25/2012] [Accepted: 03/28/2012] [Indexed: 11/15/2022]
Abstract
Humans are exposed to potentially harmful agents (bacteria, viruses, toxins) throughout our lifespan; the consequences of such exposure can alter central nervous system development. Exposure to immunogens during pregnancy increases the risk of developing neurological disorders such as schizophrenia and autism. Further, sex hormones, such as estrogen, have strong modulatory effects on immune function and have also been implicated in the development of neuropathologies (e.g., schizophrenia and depression). Similarly, animal studies have demonstrated that immunogen exposure in utero or during the neonatal period, at a time when the brain is undergoing maturation, can induce changes in learning and memory, as well as dopamine-mediated behaviors in a sex-specific manner. Literature that covers the effects of immunogens on innate immune activation and ultimately the development of the adult brain and behavior is riddled with contradictory findings, and the addition of sex as a factor only adds to the complexity. This review provides evidence that innate immune activation during critical periods of development may have effects on the adult brain in a sex-specific manner. Issues regarding sex bias in research as well as variability in animal models of immune function are discussed.
Collapse
Affiliation(s)
| | | | - Quentin J. Pittman
- Corresponding author at: Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada. Fax: +1 403 283 2700. (Q.J. Pittman)
| |
Collapse
|
44
|
Horvath B, Grasselly M, Bodecs T, Boncz I, Bodis J. Histological chorioamnionitis is associated with cerebral palsy in preterm neonates. Eur J Obstet Gynecol Reprod Biol 2012; 163:160-4. [PMID: 22682965 DOI: 10.1016/j.ejogrb.2012.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 02/13/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To determine the interaction between histological chorioamnionitis and unexplained neonatal cerebral palsy among low birth weight infants. STUDY DESIGN We studied 141 preterm infants below 1500 g delivered between 2000 and 2010. Clinical data, neonatal neuroimaging, laboratory results, the histopathological features of the placenta and gastric smear within the first hour of delivery, were evaluated. RESULTS Cerebral palsy was detected in 11 out of 141 preterm newborns (7.8%). The incidence of silent histological chorioamnionitis was 33.6% (43 of 128 cases). Chorioamniontis was significantly associated with the risk of unexplained cerebral palsy (p=0.024). There were also significant correlations between maternal genital infections and chorioamnionitis (p=0.005), and between maternal infections and a positive smear of neonatal gastric aspirates (p=0.000). The rate of cesarean section was 67.4% (95 out of 141 deliveries), and elective cesarean section was performed in 68 cases. CONCLUSION Intrauterine exposure to maternal infection was associated with a marked increase in the risk of cerebral palsy in preterm infants.
Collapse
|
45
|
von Ehrenstein OS, Neta GI, Andrews W, Goldenberg R, Goepfert A, Zhang J. Child intellectual development in relation to cytokine levels in umbilical cord blood. Am J Epidemiol 2012; 175:1191-9. [PMID: 22508393 DOI: 10.1093/aje/kwr393] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Although cytokines play a dual role in the developing neurologic system and in prenatal immune reactions, relations between fetal cytokine levels and child intellectual development remain unknown. The authors investigated associations between umbilical cord serum cytokine concentrations and intellectual outcomes in 369 children within a prospective cohort study, the Eunice Kennedy Shriver National Institute of Child Health and Human Development-University of Alabama Infant Growth Study (1985-1988). Concentrations of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interleukins 4, 10, and 12p70 were determined. The Wechsler Preschool and Primary Scale of Intelligence-Revised was administered at age 5 years, producing verbal and performance intelligence quotients (VIQ and PIQ); associations with each cytokine were evaluated using linear and logistic regression. Log-unit increases in IFN-γ (adjusted odds ratio (aOR) = 0.67, 95% confidence interval (CI): 0.46, 0.98) and interleukin-12p70 (aOR = 0.43, 95% CI: 0.21, 0.87) were inversely associated with low PIQ (score <70). One log-unit increase in TNF-α was associated with a reduced odds ratio for low VIQ (score <70) among preterm children (aOR = 0.11, 95% CI: 0.01, 0.94) and an elevated odds ratio for low VIQ among small-for-gestational-age children (aOR = 3.96, 95% CI: 0.99, 15.9). IFN-γ, which is involved in neurogenesis and perinatal adaptive immunity, may be related to fetal neurologic development overall, while TNF-α may be a marker of intellectual development in vulnerable subgroups.
Collapse
Affiliation(s)
- Ondine S von Ehrenstein
- Department of Community Health Sciences, School of Public Health, University of California, Los Angeles, 90095-1772, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Crampton SJ, Collins LM, Toulouse A, Nolan YM, O'Keeffe GW. Exposure of foetal neural progenitor cells to IL-1β impairs their proliferation and alters their differentiation - a role for maternal inflammation? J Neurochem 2012; 120:964-73. [PMID: 22192001 DOI: 10.1111/j.1471-4159.2011.07634.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
During pregnancy, activation of the maternal immune system results in inflammation in the foetal nervous system. The causative agents are pro-inflammatory cytokines like interleukin-1β (IL-1β), produced by the foetus. In this study, we examine the effect of IL-1β on the proliferation and differentiation of neural progenitor cells (NPCs) to better understand its potential effects on the developing brain. We find that the IL-1β receptor (IL-1R1) is expressed in the ventral mesencephalon of the developing brain. Furthermore, IL-1R1 is expressed on Nestin-positive, Sox-2-positive NPCs. IL-1β treatment reduced the numbers of proliferating NPCs, an effect prevented by the IL-1R1 receptor antagonist. LDH and MTT assays, and western blot analysis for cleaved caspase 3 and poly(ADP-ribose) polymerase, confirmed that this was not due to an increase in cell death but rather an induction of differentiation. To further study the effects of IL-1β on cell fate determination, we differentiated NPCs in the presence and absence of IL-1β. Il-1β promoted gliogenesis and inhibited neurogenesis, an effect that required p38-MAPK kinase signalling. In summary, these data show that exposure of NPCs to IL-1β affects their development. This necessitates an examination of the consequences that maternal immune system activation during pregnancy has on the cellular architecture of the developing brain.
Collapse
Affiliation(s)
- Sean J Crampton
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | |
Collapse
|
47
|
Harry GJ, Kraft AD. Microglia in the developing brain: a potential target with lifetime effects. Neurotoxicology 2012; 33:191-206. [PMID: 22322212 DOI: 10.1016/j.neuro.2012.01.012] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 12/15/2022]
Abstract
Microglia are a heterogenous group of monocyte-derived cells serving multiple roles within the brain, many of which are associated with immune and macrophage like properties. These cells are known to serve a critical role during brain injury and to maintain homeostasis; yet, their defined roles during development have yet to be elucidated. Microglial actions appear to influence events associated with neuronal proliferation and differentiation during development, as well as, contribute to processes associated with the removal of dying neurons or cellular debris and management of synaptic connections. These long-lived cells display changes during injury and with aging that are critical to the maintenance of the neuronal environment over the lifespan of the organism. These processes may be altered by changes in the colonization of the brain or by inflammatory events during development. This review addresses the role of microglia during brain development, both structurally and functionally, as well as the inherent vulnerability of the developing nervous system. A framework is presented considering microglia as a critical nervous system-specific cell that can influence multiple aspects of brain development (e.g., vascularization, synaptogenesis, and myelination) and have a long term impact on the functional vulnerability of the nervous system to a subsequent insult, whether environmental, physical, age-related, or disease-related.
Collapse
Affiliation(s)
- G Jean Harry
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
48
|
Prenatal exposure to lipopolysaccharide results in neurodevelopmental damage that is ameliorated by zinc in mice. Brain Behav Immun 2012; 26:326-36. [PMID: 22024135 DOI: 10.1016/j.bbi.2011.10.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/22/2011] [Accepted: 10/09/2011] [Indexed: 01/24/2023] Open
Abstract
There is converging evidence that during pregnancy a maternal immune response to infection can cause neurodevelopmental damage. Lipopolysaccharide (LPS)-mediated induction of metallothionein (MT) and subsequent hypozincaemia has been linked to fetal brain damage. Our group has demonstrated that Zn, when co-administered with LPS in early pregnancy in mice (gestation day (GD) 8), prevents fetal malformations and neurodevelopmental deficits in offspring. Others demonstrating fetal brain lesions have administered LPS much later in gestation (after GD 16), when the influence of LPS-mediated MT-induction on maternal plasma Zn levels, and the effect of Zn co-administration with LPS, are unknown. The aims of this study are firstly to examine whether LPS causes MT induction and maternal hypozincaemia in mid-to-late pregnancy, and secondly to determine if histochemical markers of inflammatory damage in fetal brain are affected by LPS and whether this damage can be alleviated with Zn treatment. Pregnant mice were injected with LPS (5 mg/kgbodywt.) or saline vehicle on GD 16 and then humanely killed at 8, 16 and 24 h for Zn and MT measurements, or concomitantly injected subcutaneously with Zn (2 mg/kgbodywt.) or saline and then killed on GD 18 and immunohistochemistry performed on fetal brain. Maternal hepatic MT was markedly induced after LPS-challenge and this was associated with a 38% reduction in maternal plasma Zn concentrations. Coincidentally, the fetuses of LPS-treated dams showed astrogliosis, extensive cell death and an increased number of cells producing TNF-α which was prevented with concomitant Zn treatment. These results support the premise that in mid-to-late pregnancy, an infection-mediated activation of a maternal immune response can cause MT induction that redistributes Zn in the mother, restricting fetal Zn supply, causing neurodevelopmental damage.
Collapse
|
49
|
Flores-Herrera H, García-López G, Díaz NF, Molina-Hernández A, Osorio-Caballero M, Soriano-Becerril D, Zaga-Clavellina V. An experimental mixed bacterial infection induced differential secretion of proinflammatory cytokines (IL-1β, TNFα) and proMMP-9 in human fetal membranes. Placenta 2012; 33:271-7. [PMID: 22280559 DOI: 10.1016/j.placenta.2012.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 12/29/2011] [Accepted: 01/03/2012] [Indexed: 11/26/2022]
Abstract
Overall, 1-4% of all births in the US are complicated by choriamnionitis. Choriamnionitis is a polymicrobial infection most often due to ascending genital microbes which, in over 65% of positive amniotic fluid cultures, involves two or more organisms. In this study, we determine the cytokines expression (IL-1β, TNFα) and prometalloproteinase activation (proMMP-2 and proMMP-9) after double o single infection an in vitro model of human fetal membranes. Fetal membranes at term were mounted in the Transwell culture system and after 24 h of infection, choriodecidual, and amnion media was collected. IL-1β and TNFα were evaluated by ELISA, whereas proMMP-9 and proMMP-2 were determined by substrate gel zymography. The choriodecidual and amnion compartments actively respond to the infectious process, which induced the secretion of IL-1β, TNFα, and proMMP-9 after either mixed or single infection. The proMMP-2 secretion profile was the same after all experimental conditions. There was no synergy between Streptococcus agalactiae and Escherichia coli for inducing the secretion of inflammatory factors or degradative metalloproteinase. In conclusion, these two bacteria could initiate different pathways to induce chorioamnioitis.
Collapse
Affiliation(s)
- H Flores-Herrera
- Department of Biochemistry and Molecular Biology, National Institute of Perinatology Isidro Espinosa de los Reyes, Montes Urales # 800, Col. Lomas de Virreyes cp 11000, Mexico City, Mexico.
| | | | | | | | | | | | | |
Collapse
|
50
|
Kapitanović Vidak H, Catela Ivković T, Jokić M, Spaventi R, Kapitanović S. The association between proinflammatory cytokine polymorphisms and cerebral palsy in very preterm infants. Cytokine 2012; 58:57-64. [PMID: 22266275 DOI: 10.1016/j.cyto.2011.12.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 11/20/2011] [Accepted: 12/24/2011] [Indexed: 12/14/2022]
Abstract
Cerebral palsy (CP) is a nonprogressive motor disorder caused by white matter damage in the developing brain and is often accompanied with cognitive and sensory disabilities. The risk of CP is higher among infants born preterm than in more mature infants. Intrauterine infection/inflammation, activation of the cytokine network and elevated levels of proinflammatory cytokines in neonatal blood or in amniotic fluid to which the preterm infant is exposed, has been identified as the most common cause of preterm delivery, periventricular leukomalacia (PVL) and CP. The aim of our study was to evaluate the possible association of four TNFα promoter single nucleotide polymorphisms (SNPs) (-1031 T/C, -857 C/T, -308 G/A and -238 G/A), two IL1β SNPs (-511 C/T and +3954 C/T) and one IL6 (-174 C/G) polymorphism with susceptibility to CP in very preterm infants. Statistically significant association between TNFα -1031 T/C high expression genotypes (TC and CC) (OR, 2.339; p=0.016) as well as between TNFα -1031 C high expression allele (OR, 2.065; p=0.013) and risk of CP was observed. In addition, statistically significant association was found between TNFα TC, CC, GG, GG -1031/-857/-308/-238 genotypes combination (OR, 3.286; p=0.034) and risk of CP. Statistically significant association between IL1β TT, CC -511/+3954 genotypes combination and risk of CP (OR, 4.000; p=0.027) was also found. In CP patients with cystic PVL (cPVL) statistically significant association was found between TNFα -1031 T/C high expression genotypes (TC and CC) (OR, 2.361; p=0.038), IL1β -511 C/T high expression genotype TT (OR, 3.215; p=0.030) as well as IL1β -511 T high expression allele (OR, 1.956; p=0.019) and risk of CP. Statistically significant association was also found in patients with cPVL between TNFα TC, CC, GG, GG -1031/-857/-308/-238 genotypes combination (OR, 4.107; p=0.024), as well as IL1β TT, CC -511/+3954 genotypes combination (OR, 7.333; p=0.005) and risk of CP. Our results suggest the role of TNFα and IL1β polymorphisms which have previously been associated with higher circulating levels of these cytokines in genetic susceptibility to white matter damage and consequently CP in very preterm infants.
Collapse
Affiliation(s)
- Helena Kapitanović Vidak
- Special Hospital for Children with Neurodevelopmental and Motor Difficulties, Goljak 2, Zagreb, Croatia
| | | | | | | | | |
Collapse
|