1
|
Lai T, Mao A, Yang L, Ren Y, Yang X, Song W, Luo Y. Association of maternal metabolic risk factors with offspring body mass index (BMI) trajectories in early childhood: a retrospective cohort study. BMJ Open 2025; 15:e088641. [PMID: 40082003 PMCID: PMC11907022 DOI: 10.1136/bmjopen-2024-088641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE This study aimed to identify body mass index (BMI) growth trajectories from birth to 24 months of age and examine the independent and additive effects of four maternal metabolic risk factors, namely prepregnancy BMI, the rate of gestational weight gain, gestational diabetes mellitus (GDM) and gestational hypertension, on offspring growth trajectories in childhood in China. DESIGN A retrospective cohort study was conducted. SETTING The study used Maternal and Child Health Management Database in Chengdu, China, including the mothers' antenatal care data, birth certificate records and 0-3-year-old children's healthcare data. PARTICIPANTS The study included mothers who gave birth between January 2014 and December 2014, and followed their offspring through 31 December 2016. The final analysis included 4492 mother-child pairs. PRIMARY OUTCOME MEASURES The primary outcomes were children's BMI measurements from birth to 24 months of age. We performed group-based trajectories modelling to identify children's BMI growth trajectories. Then, we applied logistic regression to examine the associations between maternal metabolic risk factors and offspring BMI trajectories in childhood. RESULTS Four distinct trajectories were identified: stable low (16.83%), stable average (40.69%), stable high (32.06%) and early increase (10.42%) trajectories. Relative to the stable average trajectory, maternal prepregnancy overweight (adjusted OR (aOR)=2.001, 95% CI 1.482-2.702, p<0.001), an excessive rate of gestational weight gain (aOR=1.496, 95% CI 1.138-1.966, p=0.004) and GDM (aOR=1.470, 95% CI 1.097-1.970, p=0.010) were positively associated with their offspring being in the early increase trajectory. In addition, the children's risk of being included in the early increase trajectory showed an increasing trend with an increasing number of adverse maternal metabolic risk factors. CONCLUSION Exposure to maternal prepregnancy overweight, excessive rate of weight gain and GDM resulted in a greater risk of offspring exhibiting an early increase trajectory for BMI. Decreasing maternal metabolic risk before and during pregnancy and monitoring childhood growth trajectories may prevent or delay the onset of childhood obesity.
Collapse
Affiliation(s)
- Ting Lai
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Ang Mao
- Department of Medical Administration, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Liu Yang
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Yan Ren
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Xiao Yang
- Department of Obstetrics, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Wei Song
- Department of Women and Children Health, Chengdu Municipal Health Commission, Chengdu, Sichuan, China
| | - Yingjuan Luo
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Samà M, Musillo C, Cirulli F. Counteracting the effects of maternal obesity on offspring neurodevelopment through Omega-3-based nutritional strategies. Neuroscience 2025; 566:142-148. [PMID: 39722288 DOI: 10.1016/j.neuroscience.2024.12.041] [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: 10/01/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
It is becoming increasingly recognized that, in addition to psychological stress, unbalanced maternal nutritional habits can threaten fetal brain development. Maternal obesity is one of the most pressing public health problems facing the world today, as about 40% of pregnant women are obese or gain excessive weight worldwide. This condition can negatively impact offspring's brain development, increasing the risk for autism spectrum disorders, cognitive deficits, attention deficit hyperactivity disorder, as well as anxiety and depression. In the context of fetal development, nutritional interventions may represent a feasible and safe approach for preventing the negative effects of maternal obesity. We argue that maternal Omega-3 supplementation, among the many dietary strategies available, is especially promising as it buffers oxidative stress and inflammation, both recognized as candidate mechanisms underlying the negative long-term effects of maternal obesity on the offspring. Notwithstanding the current knowledge, both preclinical studies and clinical trials are needed to refine current strategies addressing dietary content and length of administration according to individual characteristics and needs.
Collapse
Affiliation(s)
- Marianna Samà
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy.
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy
| |
Collapse
|
3
|
Barchitta M, Maugeri A, La Mastra C, Favara G, La Rosa MC, Magnano San Lio R, Gholizade Atani Y, Gallo G, Agodi A. Pre-pregnancy BMI, gestational weight gain, and telomere length in amniotic fluid: a causal graph analysis. Sci Rep 2024; 14:23396. [PMID: 39379607 PMCID: PMC11461511 DOI: 10.1038/s41598-024-74765-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
Previous investigations have suggested a potential association between pre-pregnancy body mass index (BMI) and gestational weight gain (GWG) with telomere length (TL) in various tissues of pregnant women and newborns. Nonetheless, as association does not imply causation, our objective was to investigate the causal connections among pre-pregnancy BMI, GWG, and TL in amniotic fluid. The analysis included 136 mother-child pairs from the Mamma & Bambino cohort, and three causal graph models were developed to depict the interconnections between pre-pregnancy BMI, GWG, and TL. Causal graph analysis was conducted utilizing the do-operator to estimate the causal effect of GWG and the controlled direct effect of pregestational BMI. We revealed that transitioning from non-adequate to adequate GWG had a positive impact on the probability of having "long" TL (i.e., a value greater than the population median) in all three models. When considering the effect of pre-pregnancy BMI, the highest probability of "long" TL was observed in normal weight women with adequate GWG. In contrast, the effect of adequate GWG became minimal among overweight women. These results shed light on the potential causality between pre-pregnancy BMI, GWG, and TL in amniotic fluid, emphasizing the importance of appropriate weight management before and during pregnancy for optimal TL outcomes.
Collapse
Affiliation(s)
- M Barchitta
- Department of Medical and Surgical Sciences and Advanced Technologies 'GF Ingrassia', University of Catania, Catania, Italy
| | - A Maugeri
- Department of Medical and Surgical Sciences and Advanced Technologies 'GF Ingrassia', University of Catania, Catania, Italy
| | - C La Mastra
- Department of Medical and Surgical Sciences and Advanced Technologies 'GF Ingrassia', University of Catania, Catania, Italy
| | - G Favara
- Department of Medical and Surgical Sciences and Advanced Technologies 'GF Ingrassia', University of Catania, Catania, Italy
| | - M C La Rosa
- Department of Medical and Surgical Sciences and Advanced Technologies 'GF Ingrassia', University of Catania, Catania, Italy
| | - R Magnano San Lio
- Department of Medical and Surgical Sciences and Advanced Technologies 'GF Ingrassia', University of Catania, Catania, Italy
| | - Y Gholizade Atani
- Department of Mathematics and Informatics, University of Catania, Catania, Italy
| | - G Gallo
- Department of Mathematics and Informatics, University of Catania, Catania, Italy
| | - A Agodi
- Department of Medical and Surgical Sciences and Advanced Technologies 'GF Ingrassia', University of Catania, Catania, Italy.
| |
Collapse
|
4
|
Mareboina M, Deng E, Mouratidis I, Yee NS, Pitteloud N, Georgakopoulos-Soares I, Chartoumpekis DV. A review on cell-free RNA profiling: Insights into metabolic diseases and predictive value for bariatric surgery outcomes. Mol Metab 2024; 87:101987. [PMID: 38977131 PMCID: PMC11305000 DOI: 10.1016/j.molmet.2024.101987] [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: 04/28/2024] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND The advent of liquid biopsies presents a novel, minimally invasive methodology for the detection of disease biomarkers, offering a significant advantage over traditional biopsy techniques. Particularly, the analysis of cell-free RNA (cfRNA) has garnered interest due to its dynamic expression profiles and the capability to study various RNA species, including messenger RNA (mRNA) and long non-coding RNA (lncRNA). These attributes position cfRNA as a versatile biomarker with broad potential applications in clinical research and diagnostics. SCOPE OF REVIEW This review delves into the utility of cfRNA biomarkers as prognostic tools for obesity-related comorbidities, such as diabetes, dyslipidemia, and non-alcoholic fatty liver disease. MAJOR CONCLUSIONS We evaluate the efficacy of cfRNA in forecasting metabolic outcomes associated with obesity and in identifying patients likely to experience favorable clinical outcomes following bariatric surgery. Additionally, this review synthesizes evidence from studies examining circulating cfRNA across different physiological and pathological states, with a focus on its role in diabetes, including disease progression monitoring and treatment efficacy assessment. Through this exploration, we underscore the emerging relevance of cfRNA signatures in the context of obesity and its comorbidities, setting the stage for future investigative efforts in this rapidly advancing domain.
Collapse
Affiliation(s)
- Manvita Mareboina
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Elen Deng
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ioannis Mouratidis
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Nelson S Yee
- Division of Hematology-Oncology, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Next-Generation Therapies Program, Penn State Cancer Institute, Hershey, PA, USA
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Dionysios V Chartoumpekis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
5
|
Olga L, Sovio U, Wong H, Smith GCS, Aiken CEM. Maternal high body mass index, but not gestational diabetes, is associated with poorer educational attainment in mid-childhood. Am J Obstet Gynecol 2024; 231:120.e1-120.e9. [PMID: 37981092 DOI: 10.1016/j.ajog.2023.11.1227] [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: 05/05/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Previous studies suggest that gestational diabetes mellitus is associated with poorer cognitive outcomes in children. However, confounding factors, especially maternal body mass index, have been poorly accounted for. OBJECTIVE This study aimed to examine the independent associations between maternal body mass index, gestational diabetes mellitus status, and educational outcomes. STUDY DESIGN Antenatal data from a prospective birth cohort (Pregnancy Outcome Prediction Study, 2008-2012, Cambridge, United Kingdom) were linked to mid-childhood educational outcomes (Department for Education, United Kingdom). A total of 3249 children born at term were stratified by maternal gestational diabetes mellitus status and body mass index at booking (<25 vs ≥25 kg/m2). Regression models adjusted for relevant maternal, child, and socioeconomic factors were used to determine associations with academic outcomes at ages of 5 to 7 years. RESULTS No differences in educational attainment were found between children exposed to gestational diabetes mellitus and nonexposed children. Neither maternal glucose levels measured at 11 to 14 or 24 to 28 weeks, nor acceleration of the fetal abdominal circumference growth velocity were related to educational attainment at ages of 5 to 7 years. Children of mothers with booking body mass index ≥25 kg/m2 (vs <25 kg/m2) were ∼50% more likely to not meet expected educational standards regardless of gestational diabetes mellitus status (age 5: adjusted odds ratio, 1.44; 95% confidence interval, 1.19-1.74; P<.001; age 6: adjusted odds ratio, 1.61; 95% confidence interval, 1.28-2.02; P<.001). The association between maternal body mass index and offspring educational attainment is dose-dependent and robust to stratification by gestational diabetes mellitus status and adjustment for socioeconomic factors. CONCLUSION Mid-childhood educational attainment is not associated with maternal glucose status. This may provide important reassurance for pregnant women and clinicians. However, maternal body mass index is associated with lower childhood educational attainment and may be modifiable with intervention before or during pregnancy.
Collapse
Affiliation(s)
- Laurentya Olga
- Department of Obstetrics and Gynaecology, Rosie Hospital, National Institute for Health and Care Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ulla Sovio
- Department of Obstetrics and Gynaecology, Rosie Hospital, National Institute for Health and Care Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Hilary Wong
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, Rosie Hospital, National Institute for Health and Care Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Catherine E M Aiken
- Department of Obstetrics and Gynaecology, Rosie Hospital, National Institute for Health and Care Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
6
|
Akgun Aktaş B, Ecem K, Murat H, Gökçe Ozcan K, Ozgür K, Atakan T, Dilek S. Fetal cortical development and neurosonographic findings in obese pregnant women: a case control study from a tertiary hospital. Arch Gynecol Obstet 2024; 310:83-91. [PMID: 37831178 DOI: 10.1007/s00404-023-07258-4] [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: 08/10/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
PURPOSE To investigate the effect of obesity on fetal cortical development. METHODS This prospective cross-sectional study was conducted with 91 pregnant women. Fetal neurosonography scans were performed in the third trimester, and according to body mass index (BMI) values, the patients were evaluated in two groups: obese (BMI ≥ 30 kg/m2) and normal weight (BMI < 30 kg/m2). During neurosonography, fetal insular depth and Sylvian fissures, parieo-occipital and cingulate fissure depth, frontal lobe length, and the sizes of the corpus callosum and cavum septum pellucidum were measured using a transvaginal approach. Fetal cortical development and Sylvian fissure operculization were graded. RESULTS The number of patients with grade 2 fetal cortical development was significantly higher among the pregnant women in the obese group compared to the normal weight group (n = 17, 41.5% and n = 8, 16.0%, respectively; p = 0.007). In the obese group, the number of pregnant women with grade 4 or below fetal Sylvian fissure operculization was significantly higher (n = 13, 31.7%), and the number of those with grade 9 or above operculization was significantly lower (n = 1, 2.5%) (p = 0.003). The fetal insular depth, frontal lobe anterior-posterior diameter, cingulate fissure depth, and corpus callosum thickness were lower in the obese group, albeit with no significant difference. Parieto-occipital depth significantly decreased in the obese group [6.8 (6) mm)] compared to the control group [10.5 (7.2) mm)] (p = 0.008). The fetal Sylvian fissure ratio and the cavum septum pellucidum ratio were found to be similar between the groups. CONCLUSION The data obtained from this study showed that obesity caused fetal cortical changes in pregnant women.
Collapse
Affiliation(s)
- Betül Akgun Aktaş
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey.
| | - Kaya Ecem
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
| | - Haksever Murat
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
| | - Kılınç Gökçe Ozcan
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
| | - Kara Ozgür
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
- Department of Obstetrics and Gynecology, University of Health Sciences, Istanbul, Turkey
| | - Tanacan Atakan
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
- Department of Obstetrics and Gynecology, University of Health Sciences, Istanbul, Turkey
| | - Sahin Dilek
- Department of Obstetrics and Gynecology, Ministry of Health, Ankara City Hospital, Üniversiteler Mahallesi 1604. Cadde No: 9 Çankaya, Ankara, Turkey
- Department of Obstetrics and Gynecology, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
7
|
Merabova N, Ugartemendia L, Edlow AG, Ibarra C, Darbinian N, Tatevosian G, Goetzl L. Maternal obesity: sex-specific in utero changes in fetal brain autophagy and mTOR. Obesity (Silver Spring) 2024; 32:1136-1143. [PMID: 38644654 DOI: 10.1002/oby.24017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 04/23/2024]
Abstract
OBJECTIVE Maternal obesity affects 39.7% of reproductive-age women in the United States. Emerging research has suggested that in utero exposure to maternal obesity is associated with adverse neurodevelopmental outcomes, but knowledge of underlying mechanisms in human samples is lacking. METHODS A matched case-control study was performed in women with singleton fetuses who were undergoing elective pregnancy termination at gestational ages 15 to 21 weeks. Maternal adiponectin levels from plasma were measured using ELISA kits. RNA was extracted from fetal brain tissue using RNeasy Mini Kit (QIAGEN). mRNA expression from ADIPOR1, ADIPOR2, MTOR, ATG5, ATG7, BECN1, and MAP1LC3B was quantified through the ΔΔCt method and using GAPDH as a housekeeping gene. RESULTS We have identified transcription patterns associated with inhibition of autophagy in male fetal brain tissue exposed to maternal obesity (↑MTOR, ↓ATG5, ↓ATG7, and ↓MAP1LC3B), with female fetuses demonstrating either no change in transcription or nonsignificant changes associated with increased autophagy. There was significant downregulation of the autophagy-associated gene BECN1 in both male and female individuals who were exposed to obesity in utero. CONCLUSIONS We present novel evidence suggesting that in utero exposure to maternal obesity in humans may significantly affect neurodevelopment, especially in male fetuses, through alterations in normal autophagy molecular mechanisms and with adiponectin as a potential mediator.
Collapse
Affiliation(s)
- Nana Merabova
- Department of Family Medicine, Medical College of Wisconsin-Prevea, Green Bay, Wisconsin, USA
| | - Lierni Ugartemendia
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, Massachusetts, USA
| | - Claudia Ibarra
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nune Darbinian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania, USA
| | - Gabriel Tatevosian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania, USA
| | - Laura Goetzl
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
8
|
del Valle E, Rubio-Sardón N, Menéndez-Pérez C, Martínez-Pinilla E, Navarro A. Apolipoprotein D as a Potential Biomarker in Neuropsychiatric Disorders. Int J Mol Sci 2023; 24:15631. [PMID: 37958618 PMCID: PMC10650001 DOI: 10.3390/ijms242115631] [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: 10/08/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Neuropsychiatric disorders (NDs) are a diverse group of pathologies, including schizophrenia or bipolar disorders, that directly affect the mental and physical health of those who suffer from them, with an incidence that is increasing worldwide. Most NDs result from a complex interaction of multiple genes and environmental factors such as stress or traumatic events, including the recent Coronavirus Disease (COVID-19) pandemic. In addition to diverse clinical presentations, these diseases are heterogeneous in their pathogenesis, brain regions affected, and clinical symptoms, making diagnosis difficult. Therefore, finding new biomarkers is essential for the detection, prognosis, response prediction, and development of new treatments for NDs. Among the most promising candidates is the apolipoprotein D (Apo D), a component of lipoproteins implicated in lipid metabolism. Evidence suggests an increase in Apo D expression in association with aging and in the presence of neuropathological processes. As a part of the cellular neuroprotective defense machinery against oxidative stress and inflammation, changes in Apo D levels have been demonstrated in neuropsychiatric conditions like schizophrenia (SZ) or bipolar disorders (BPD), not only in some brain areas but in corporal fluids, i.e., blood or serum of patients. What is not clear is whether variation in Apo D quantity could be used as an indicator to detect NDs and their progression. This review aims to provide an updated view of the clinical potential of Apo D as a possible biomarker for NDs.
Collapse
Affiliation(s)
- Eva del Valle
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Nuria Rubio-Sardón
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Carlota Menéndez-Pérez
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Eva Martínez-Pinilla
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Ana Navarro
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| |
Collapse
|
9
|
Zhao D, Liu Y, Jia S, He Y, Wei X, Liu D, Ma W, Luo W, Gu H, Yuan Z. Influence of maternal obesity on the multi-omics profiles of the maternal body, gestational tissue, and offspring. Biomed Pharmacother 2022; 151:113103. [PMID: 35605294 DOI: 10.1016/j.biopha.2022.113103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022] Open
Abstract
Epidemiological studies show that obesity during pregnancy affects more than half of the pregnancies in the developed countries and is associated with obstetric problems and poor outcomes. Obesity tends to increase the incidence of complications. Furthermore, the resulting offspring are also adversely affected. However, the molecular mechanisms of obesity leading to poor pregnancy outcomes remain unclear. Omics methods are used for genetic diagnosis and marker discovery. The aim of this review was to summarize the maternal and fetal pathophysiological alterations induced by gestational obesity,identified using multi-omics detection techniques, and to generalize the biological functions and potential mechanisms of the differentially expressed molecules.
Collapse
Affiliation(s)
- Duan Zhao
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Yusi Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Shanshan Jia
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Yiwen He
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Xiaowei Wei
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Wei Ma
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| |
Collapse
|
10
|
Liu P, Vossaert L. Emerging technologies for prenatal diagnosis: The application of whole genome and RNA sequencing. Prenat Diagn 2022; 42:686-696. [PMID: 35416301 PMCID: PMC10014115 DOI: 10.1002/pd.6146] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022]
Abstract
DNA sequencing technologies for clinical genetic testing have been rapidly evolving in recent years, and steadily become more important within the field of prenatal diagnostics. This review aims to give an overview of recent developments and to describe how they have the potential to fill the gaps of the currently clinically implemented methods for prenatal diagnosis of various genetic disorders. It has been shown for postnatal testing that whole genome sequencing provides a set of added benefits compared to exome sequencing, and it is to be expected that this will be the case for prenatal testing as well. RNA-sequencing, already used postnatally, can provide valuable complementary data to DNA-based testing, and aid in variant interpretation. While not ready for clinical implementation, emerging technologies such as long-read and Hi-C sequencing analyses might add to the toolbox for interpreting the expanding genetic data sets generated by genome-wide sequencing. Lastly, we also discuss some more practical implications of introducing these emerging technologies, which generate larger and larger genomic data sets, in the prenatal field.
Collapse
Affiliation(s)
- Pengfei Liu
- Baylor College of Medicine and Baylor Genetics, Houston, Texas, USA
| | | |
Collapse
|
11
|
The Relationship between Telomere Length and Gestational Weight Gain: Findings from the Mamma & Bambino Cohort. Biomedicines 2021; 10:biomedicines10010067. [PMID: 35052747 PMCID: PMC8773008 DOI: 10.3390/biomedicines10010067] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/21/2022] Open
Abstract
Inadequate gestational weight gain (GWG) affects a growing number of pregnancies, influencing intrauterine environment and long-term health. Uncovering molecular mechanisms associated with GWG could be helpful to develop public health strategies for tackling this issue. Here, our study aimed to understand the relationship of DNA telomere length with weigh gain during pregnancy, using data and samples from the ongoing prospective “Mamma & Bambino” study (Catania, Italy). GWG was calculated according to the Institute of Medicine (IOM) guidelines. Relative telomere length was assessed by real-time quantitative polymerase chain reaction in 252 samples of maternal leucocyte DNA (mlDNA) and 150 samples of cell-free DNA (cfDNA) from amniotic fluid. We observed that relative telomere length of mlDNA seemed to weakly increase with GWG. In contrast, telomere length of cfDNA exhibited a U-shaped relationship with GWG. Women with adequate GWG showed longer telomere length than those who gained weight inadequately. Accordingly, the logistic regression model confirmed the association between telomere length of cfDNA and adequate GWG, after adjusting for potential confounders. Our findings suggest an early effect of GWG on telomere length of cfDNA, which could represent a molecular mechanism underpinning the effects of maternal behaviours on foetal well-being.
Collapse
|
12
|
Soullane S, Spence AR, Abenhaim HA. Association of placental pathology and gross morphology with autism spectrum disorders. Autism Res 2021; 15:531-538. [DOI: 10.1002/aur.2658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Safiya Soullane
- Faculty of Medicine McGill University Montreal Quebec Canada
| | - Andrea R. Spence
- Centre for Clinical Epidemiology and Community Studies Jewish General Hospital Montreal Quebec Canada
| | - Haim A. Abenhaim
- Centre for Clinical Epidemiology and Community Studies Jewish General Hospital Montreal Quebec Canada
- Department of Obstetrics and Gynecology Jewish General Hospital, McGill University Montreal Quebec Canada
| |
Collapse
|
13
|
Sanchez D, Ganfornina MD. The Lipocalin Apolipoprotein D Functional Portrait: A Systematic Review. Front Physiol 2021; 12:738991. [PMID: 34690812 PMCID: PMC8530192 DOI: 10.3389/fphys.2021.738991] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022] Open
Abstract
Apolipoprotein D is a chordate gene early originated in the Lipocalin protein family. Among other features, regulation of its expression in a wide variety of disease conditions in humans, as apparently unrelated as neurodegeneration or breast cancer, have called for attention on this gene. Also, its presence in different tissues, from blood to brain, and different subcellular locations, from HDL lipoparticles to the interior of lysosomes or the surface of extracellular vesicles, poses an interesting challenge in deciphering its physiological function: Is ApoD a moonlighting protein, serving different roles in different cellular compartments, tissues, or organisms? Or does it have a unique biochemical mechanism of action that accounts for such apparently diverse roles in different physiological situations? To answer these questions, we have performed a systematic review of all primary publications where ApoD properties have been investigated in chordates. We conclude that ApoD ligand binding in the Lipocalin pocket, combined with an antioxidant activity performed at the rim of the pocket are properties sufficient to explain ApoD association with different lipid-based structures, where its physiological function is better described as lipid-management than by long-range lipid-transport. Controlling the redox state of these lipid structures in particular subcellular locations or extracellular structures, ApoD is able to modulate an enormous array of apparently diverse processes in the organism, both in health and disease. The new picture emerging from these data should help to put the physiological role of ApoD in new contexts and to inspire well-focused future research.
Collapse
Affiliation(s)
- Diego Sanchez
- Instituto de Biologia y Genetica Molecular, Unidad de Excelencia, Universidad de Valladolid-Consejo Superior de Investigaciones Cientificas, Valladolid, Spain
| | - Maria D Ganfornina
- Instituto de Biologia y Genetica Molecular, Unidad de Excelencia, Universidad de Valladolid-Consejo Superior de Investigaciones Cientificas, Valladolid, Spain
| |
Collapse
|
14
|
Adipose-Derived Lipid-Binding Proteins: The Good, the Bad and the Metabolic Diseases. Int J Mol Sci 2021; 22:ijms221910460. [PMID: 34638803 PMCID: PMC8508731 DOI: 10.3390/ijms221910460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue releases a large range of bioactive factors called adipokines, many of which are involved in inflammation, glucose homeostasis and lipid metabolism. Under pathological conditions such as obesity, most of the adipokines are upregulated and considered as deleterious, due to their pro-inflammatory, pro-atherosclerotic or pro-diabetic properties, while only a few are downregulated and would be designated as beneficial adipokines, thanks to their counteracting properties against the onset of comorbidities. This review focuses on six adipose-derived lipid-binding proteins that have emerged as key factors in the development of obesity and diabetes: Retinol binding protein 4 (RBP4), Fatty acid binding protein 4 (FABP4), Apolipoprotein D (APOD), Lipocalin-2 (LCN2), Lipocalin-14 (LCN14) and Apolipoprotein M (APOM). These proteins share structural homology and capacity to bind small hydrophobic molecules but display opposite effects on glucose and lipid metabolism. RBP4 and FABP4 are positively associated with metabolic syndrome, while APOD and LCN2 are ubiquitously expressed proteins with deleterious or beneficial effects, depending on their anatomical site of expression. LCN14 and APOM have been recently identified as adipokines associated with healthy metabolism. Recent findings on these lipid-binding proteins exhibiting detrimental or protective roles in human and murine metabolism and their involvement in metabolic diseases are also discussed.
Collapse
|
15
|
The amniotic fluid cell-free transcriptome in spontaneous preterm labor. Sci Rep 2021; 11:13481. [PMID: 34188072 PMCID: PMC8242007 DOI: 10.1038/s41598-021-92439-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/03/2021] [Indexed: 02/03/2023] Open
Abstract
The amniotic fluid (AF) cell-free RNA was shown to reflect physiological and pathological processes in pregnancy, but its value in the prediction of spontaneous preterm delivery is unknown. Herein we profiled cell-free RNA in AF samples collected from women who underwent transabdominal amniocentesis after an episode of spontaneous preterm labor and subsequently delivered within 24 h (n = 10) or later (n = 28) in gestation. Expression of known placental single-cell RNA-Seq signatures was quantified in AF cell-free RNA and compared between the groups. Random forest models were applied to predict time-to-delivery after amniocentesis. There were 2385 genes differentially expressed in AF samples of women who delivered within 24 h of amniocentesis compared to gestational age-matched samples from women who delivered after 24 h of amniocentesis. Genes with cell-free RNA changes were associated with immune and inflammatory processes related to the onset of labor, and the expression of placental single-cell RNA-Seq signatures of immune cells was increased with imminent delivery. AF transcriptomic prediction models captured these effects and predicted delivery within 24 h of amniocentesis (AUROC = 0.81). These results may inform the development of biomarkers for spontaneous preterm birth.
Collapse
|
16
|
The Amniotic Fluid Cell-Free Transcriptome Provides Novel Information about Fetal Development and Placental Cellular Dynamics. Int J Mol Sci 2021; 22:ijms22052612. [PMID: 33807645 PMCID: PMC7961801 DOI: 10.3390/ijms22052612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022] Open
Abstract
The amniotic fluid (AF) is a complex biofluid that reflects fetal well-being during development. AF con be divided into two fractions, the supernatant and amniocytes. The supernatant contains cell-free components, including placenta-derived microparticles, protein, cell-free fetal DNA, and cell-free fetal RNA from the fetus. Cell-free mRNA (cfRNA) analysis holds a special position among high-throughput analyses, such as transcriptomics, proteomics, and metabolomics, owing to its ease of profiling. The AF cell-free transcriptome differs from the amniocyte transcriptome and alters with the progression of pregnancy and is often associated with the development of various organ systems including the fetal lung, skin, brain, pancreas, adrenal gland, gastrointestinal system, etc. The AF cell-free transcriptome is affected not only by normal physiologies, such as fetal sex, gestational age, and fetal maturity, but also by pathologic mechanisms such as maternal obesity, and genetic syndromes (Down, Edward, Turner, etc.), as well as pregnancy complications (preeclampsia, intrauterine growth restriction, preterm birth, etc.). cfRNA in the amniotic fluid originates from the placenta and fetal organs directly contacting the amniotic fluid as well as from the fetal plasma across the placenta. The AF transcriptome may reflect the fetal and placental development and therefore aid in the monitoring of normal and abnormal development.
Collapse
|
17
|
Ermakov EA, Dmitrieva EM, Parshukova DA, Kazantseva DV, Vasilieva AR, Smirnova LP. Oxidative Stress-Related Mechanisms in Schizophrenia Pathogenesis and New Treatment Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8881770. [PMID: 33552387 PMCID: PMC7847339 DOI: 10.1155/2021/8881770] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/15/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023]
Abstract
Schizophrenia is recognized to be a highly heterogeneous disease at various levels, from genetics to clinical manifestations and treatment sensitivity. This heterogeneity is also reflected in the variety of oxidative stress-related mechanisms contributing to the phenotypic realization and manifestation of schizophrenia. At the molecular level, these mechanisms are supposed to include genetic causes that increase the susceptibility of individuals to oxidative stress and lead to gene expression dysregulation caused by abnormal regulation of redox-sensitive transcriptional factors, noncoding RNAs, and epigenetic mechanisms favored by environmental insults. These changes form the basis of the prooxidant state and lead to altered redox signaling related to glutathione deficiency and impaired expression and function of redox-sensitive transcriptional factors (Nrf2, NF-κB, FoxO, etc.). At the cellular level, these changes lead to mitochondrial dysfunction and metabolic abnormalities that contribute to aberrant neuronal development, abnormal myelination, neurotransmitter anomalies, and dysfunction of parvalbumin-positive interneurons. Immune dysfunction also contributes to redox imbalance. At the whole-organism level, all these mechanisms ultimately contribute to the manifestation and development of schizophrenia. In this review, we consider oxidative stress-related mechanisms and new treatment perspectives associated with the correction of redox imbalance in schizophrenia. We suggest that not only antioxidants but also redox-regulated transcription factor-targeting drugs (including Nrf2 and FoxO activators or NF-κB inhibitors) have great promise in schizophrenia. But it is necessary to develop the stratification criteria of schizophrenia patients based on oxidative stress-related markers for the administration of redox-correcting treatment.
Collapse
Affiliation(s)
- Evgeny A. Ermakov
- Laboratory of Repair Enzymes, Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Elena M. Dmitrieva
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Daria A. Parshukova
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | | | | | - Liudmila P. Smirnova
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| |
Collapse
|
18
|
Drag MH, Kilpeläinen TO. Cell-free DNA and RNA-measurement and applications in clinical diagnostics with focus on metabolic disorders. Physiol Genomics 2020; 53:33-46. [PMID: 33346689 DOI: 10.1152/physiolgenomics.00086.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Circulating cell-free DNA (cfDNA) and RNA (cfRNA) hold enormous potential as a new class of biomarkers for the development of noninvasive liquid biopsies in many diseases and conditions. In recent years, cfDNA and cfRNA have been studied intensely as tools for noninvasive prenatal testing, solid organ transplantation, cancer screening, and monitoring of tumors. In obesity, higher cfDNA concentration indicates accelerated cellular turnover of adipocytes during expansion of adipose mass and may be directly involved in the development of adipose tissue insulin resistance by inducing inflammation. Furthermore, cfDNA and cfRNA have promising diagnostic value in a range of obesity-related metabolic disorders, such as nonalcoholic fatty liver disease, type 2 diabetes, and diabetic complications. Here, we review the current and future applications of cfDNA and cfRNA within clinical diagnostics, discuss technical and analytical challenges in the field, and summarize the opportunities of using cfDNA and cfRNA in the diagnostics and prognostics of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Markus H Drag
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Del Vecchio G, Li Q, Li W, Thamotharan S, Tosevska A, Morselli M, Sung K, Janzen C, Zhou X, Pellegrini M, Devaskar SU. Cell-free DNA Methylation and Transcriptomic Signature Prediction of Pregnancies with Adverse Outcomes. Epigenetics 2020; 16:642-661. [PMID: 33045922 DOI: 10.1080/15592294.2020.1816774] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although analysis of maternal plasma cell-free content has been employed for screening of genetic abnormalities within a pregnancy, limited attention has been paid to its use for the detection of adverse pregnancy outcomes (APOs) based on placental function. Here we investigated cell-free DNA and RNA content of 102 maternal and 25 cord plasma samples. Employing a novel deconvolution methodology, we found that during the first trimester, placenta-specific DNA increased prior to the subsequent development of gestational diabetes with no change in patients with preeclampsia while decreasing with maternal obesity. Moreover, using cell-free RNA sequencing, APOs revealed 71 differentially expressed genes early in pregnancy. We noticed the upregulation of S100A8, MS4A3, and MMP8 that have been already associated with APOs but also the upregulation of BCL2L15 and the downregulation of ALPL that have never been associated with APOs. We constructed a classifier with a positive predictive ability (AUC) of 0.91 for APOs, 0.86 for preeclampsia alone and 0.64 for GDM. We conclude that placenta-specific cell-free nucleic acids during early gestation provide the possibility of predicting APOs prior to the emergence of characteristic clinical features.
Collapse
Affiliation(s)
- Giorgia Del Vecchio
- Departments of Pediatrics David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Qingjiao Li
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wenyuan Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Shanthie Thamotharan
- Departments of Pediatrics David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Anela Tosevska
- Departments of Molecular, Cellular and Developmental Biology, University of California Los Angeles, California, USA
| | - Marco Morselli
- Departments of Molecular, Cellular and Developmental Biology, University of California Los Angeles, California, USA
| | - Kyunghyun Sung
- Department of Radiological Sciences, University of California Los Angeles, California, USA
| | - Carla Janzen
- Department of Obstetrics & Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Xianghong Zhou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Matteo Pellegrini
- Departments of Molecular, Cellular and Developmental Biology, University of California Los Angeles, California, USA
| | - Sherin U Devaskar
- Departments of Pediatrics David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
20
|
Siracusano M, Riccioni A, Abate R, Benvenuto A, Curatolo P, Mazzone L. Vitamin D Deficiency and Autism Spectrum Disorder. Curr Pharm Des 2020; 26:2460-2474. [PMID: 32294031 DOI: 10.2174/1381612826666200415174311] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/08/2020] [Indexed: 12/24/2022]
Abstract
Vitamin D is a neurosteroid hormone crucially involved in neurodevelopment. Neural cell proliferation, neurotransmission, oxidative stress and immune function represent the main mechanisms mediated by vitamin D in the Central Nervous System. Therefore, its deficiency during pregnancy and early childhood may significantly impact on a developing brain, leading to possible adverse neuropsychological outcomes including Autism Spectrum Disorder (ASD). Significant vitamin D deficiency is described within children affected by ASD and in pregnant mothers whose offspring will later develop ASD, suggesting a possible role of the hormone as a contributing risk factor in the etiopathogenesis of ASD. We reviewed the actual literature on the potential contributing role of prenatal and early postnatal vitamin D deficiency in ASD etiopathogenesis, at both genetic and environmental levels, and the possible effect of vitamin D supplementation in autistic children. Conflicting but promising results emerged on the topic. Further Randomized Controlled Trials studies carried out during pregnancy and early infancy are necessary for better understanding the possible contribution of vitamin D deficiency in the etiopathogenesis of autism and the potential efficacy of the hormone supplementation in the improvement of ASD core symptoms.
Collapse
Affiliation(s)
- Martina Siracusano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.,PHD Student in Experimental Medicine- Neuroscience, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila AQ, Italy
| | - Assia Riccioni
- Child Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy
| | - Roberta Abate
- Child Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy
| | - Arianna Benvenuto
- Child Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy
| | - Luigi Mazzone
- Child Neurology and Psychiatry Unit, System Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy
| |
Collapse
|
21
|
Tarca AL, Romero R, Pique-Regi R, Pacora P, Done B, Kacerovsky M, Bhatti G, Jaiman S, Hassan SS, Hsu CD, Gomez-Lopez N. Amniotic fluid cell-free transcriptome: a glimpse into fetal development and placental cellular dynamics during normal pregnancy. BMC Med Genomics 2020; 13:25. [PMID: 32050959 PMCID: PMC7017452 DOI: 10.1186/s12920-020-0690-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background The amniotic fluid (AF) cell-free transcriptome is modulated by physiologic and pathologic processes during pregnancy. AF gene expression changes with advancing gestation reflect fetal development and organ maturation; yet, defining normal expression and splicing patterns for biomarker discovery in obstetrics requires larger heterogeneous cohorts, evaluation of potential confounding factors, and novel analytical approaches. Methods Women with a normal pregnancy who had an AF sample collected during midtrimester (n = 30) or at term gestation (n = 68) were included. Expression profiling at exon level resolution was performed using Human Transcriptome Arrays. Differential expression was based on moderated t-test adjusted p < 0.05 and fold change > 1.25; for differential splicing, a splicing index > 2 and adjusted p < 0.05 were required. Functional profiling was used to interpret differentially expressed or spliced genes. The expression of tissue-specific and cell-type specific signatures defined by single-cell genomics was quantified and correlated with covariates. In-silico validation studies were performed using publicly available datasets. Results 1) 64,071 genes were detected in AF, with 11% of the coding and 6% of the non-coding genes being differentially expressed between midtrimester and term gestation. Expression changes were highly correlated with those previously reported (R > 0.79, p < 0.001) and featured increased expression of genes specific to the trachea, salivary glands, and lung and decreased expression of genes specific to the cardiac myocytes, uterus, and fetal liver, among others. 2) Single-cell RNA-seq signatures of the cytotrophoblast, Hofbauer cells, erythrocytes, monocytes, T and B cells, among others, showed complex patterns of modulation with gestation (adjusted p < 0.05). 3) In 17% of the genes detected, we found differential splicing with advancing gestation in genes related to brain development processes and immunity pathways, including some that were missed based on differential expression analysis alone. Conclusions This represents the largest AF transcriptomics study in normal pregnancy, reporting for the first time that single-cell genomic signatures can be tracked in the AF and display complex patterns of expression during gestation. We also demonstrate a role for alternative splicing in tissue-identity acquisition, organ development, and immune processes. The results herein may have implications for the development of fetal testing to assess placental function and fetal organ maturity.
Collapse
Affiliation(s)
- Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA. .,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA.
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA. .,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA. .,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA. .,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA. .,Detroit Medical Center, Detroit, MI, USA. .,Department of Pathology, Hutzel Women's Hospital, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
| | - Marian Kacerovsky
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sunil Jaiman
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.,Department of Pathology, Hutzel Women's Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sonia S Hassan
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 𝐸𝑢𝑛𝑖𝑐𝑒 𝐾𝑒𝑛𝑛𝑒𝑑𝑦 𝑆ℎ𝑟𝑖𝑣𝑒𝑟 National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA. .,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicin, Detroit, MI, USA.
| |
Collapse
|
22
|
Cirulli F, Musillo C, Berry A. Maternal Obesity as a Risk Factor for Brain Development and Mental Health in the Offspring. Neuroscience 2020; 447:122-135. [PMID: 32032668 DOI: 10.1016/j.neuroscience.2020.01.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/30/2022]
Abstract
Maternal obesity plays a key role in the health trajectory of the offspring. Although research on this topic has largely focused on the potential of this condition to increase the risk for child obesity, it is becoming more and more evident that it can also significantly impact cognitive function and mental health. The mechanisms underlying these effects are starting to be elucidated and point to the placenta as a critical organ that may mediate changes in the response to stress, immune function and oxidative stress. Long-term effects of maternal obesity may rely upon epigenetic changes in selected genes that are involved in metabolic and trophic regulations of the brain. More recent evidence also indicates the gut microbiota as a potential mediator of these effects. Overall, understanding cause-effect relationships can allow the development of preventive measures that could rely upon dietary changes in the mother and the offspring. Addressing diets appears more feasible than developing new pharmacological targets and has the potential to affect the multiple interconnected physiological pathways engaged by these complex regulations, allowing prevention of both metabolic and mental disorders.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
23
|
Cárdenas-Tueme M, Montalvo-Martínez L, Maldonado-Ruiz R, Camacho-Morales A, Reséndez-Pérez D. Neurodegenerative Susceptibility During Maternal Nutritional Programing: Are Central and Peripheral Innate Immune Training Relevant? Front Neurosci 2020; 14:13. [PMID: 32116490 PMCID: PMC7010854 DOI: 10.3389/fnins.2020.00013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/08/2020] [Indexed: 12/24/2022] Open
Abstract
Maternal overnutrition modulates body weight, development of metabolic failure and, potentially, neurodegenerative susceptibility in the offspring. Overnutrition sets a chronic pro-inflammatory profile that integrates peripheral and central immune activation nodes, damaging neuronal physiology and survival. Innate immune cells exposed to hypercaloric diets might experience trained immunity. Here, we address the role of maternal overnutrition as a trigger for central and peripheral immune training and its contribution to neurodegeneration and the molecular nodes implicated in the Nod-like receptor protein 3 (NLRP3) inflammasome pathway leading to immune training. We propose that maternal overnutrition leads to peripheral or central immune training that favor neurodegenerative susceptibility in the offspring.
Collapse
Affiliation(s)
- Marcela Cárdenas-Tueme
- Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Larisa Montalvo-Martínez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Roger Maldonado-Ruiz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Alberto Camacho-Morales
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- Centro de Investigación y Desarrollo en Ciencias de la Salud, Unidad de Neurometabolismo, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Diana Reséndez-Pérez
- Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| |
Collapse
|
24
|
Jasinska AJ, Rostamian D, Davis AT, Kavanagh K. Transcriptomic Analysis of Cell-free Fetal RNA in the Amniotic Fluid of Vervet Monkeys ( Chlorocebus sabaeus). Comp Med 2020; 70:67-74. [PMID: 31969210 PMCID: PMC7024774 DOI: 10.30802/aalas-cm-19-000037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/22/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022]
Abstract
NHP are important translational models for understanding the genomic underpinnings of growth, development, fetal programming, and predisposition to disease, with potential for the development of early health biomarkers. Understanding how prenatal gene expression is linked to pre- and postnatal health and development requires methods for assessing the fetal transcriptome. Here we used RNAseq methodology to analyze the expression of cell-free fetal RNA in the amniotic fluid supernatant (AFS) of vervet monkeys. Despite the naturally high level of degradation of free-floating RNA, we detected more than 10,000 gene transcripts in vervet AFS. The most highly expressed genes were H19, IGF2, and TPT1, which are involved in embryonic growth and glycemic health. We noted global similarities in expression profiles between vervets and humans, with genes involved in embryonic growth and glycemic health among the genes most highly expressed in AFS. Our study demonstrates both the feasibility and usefulness of prenatal transcriptomic profiles, by using amniocentesis procedures to obtain AFS and cell-free fetal RNA from pregnant vervets.
Collapse
Affiliation(s)
- Anna J Jasinska
- Center for Neurobehavioral Genetics, University of California-Los Angeles, Los Angeles, California; Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland;,
| | - Dalar Rostamian
- Center for Neurobehavioral Genetics, University of California-Los Angeles, Los Angeles, California
| | - Ashley T Davis
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Kylie Kavanagh
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; Department of Biomedicine, University of Tasmania, Hobart, Australia
| |
Collapse
|
25
|
Nichols AR, Rundle AG, Factor-Litvak P, Insel BJ, Hoepner L, Rauh V, Perera F, Widen EM. Prepregnancy obesity is associated with lower psychomotor development scores in boys at age 3 in a low-income, minority birth cohort. J Dev Orig Health Dis 2020; 11:49-57. [PMID: 31486358 PMCID: PMC6934918 DOI: 10.1017/s2040174419000412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Whether maternal obesity and gestational weight gain (GWG) are associated with early-childhood development in low-income, urban, minority populations, and whether effects differ by child sex remain unknown. This study examined the impact of prepregnancy BMI and GWG on early childhood neurodevelopment in the Columbia Center for Children's Environmental Health Mothers and Newborns study. Maternal prepregnancy weight was obtained by self-report, and GWG was assessed from participant medical charts. At child age 3 years, the Psychomotor Development Index (PDI) and Mental Development Index (MDI) of the Bayley Scales of Infant Intelligence were completed. Sex-stratified linear regression models assessed associations between prepregnancy BMI and pregnancy weight gain z-scores with child PDI and MDI scores, adjusting for covariates. Of 382 women, 48.2% were normal weight before pregnancy, 24.1% overweight, 23.0% obese, and 4.7% underweight. At 3 years, mean scores on the PDI and MDI were higher among girls compared to boys (PDI: 102.3 vs. 97.2, P = 0.0002; MDI: 92.8 vs. 88.3, P = 0.0001). In covariate-adjusted models, maternal obesity was markedly associated with lower PDI scores in boys [b = -7.81, 95% CI: (-13.08, -2.55), P = 0.004], but not girls. Maternal BMI was not associated with MDI in girls or boys, and GWG was not associated with PDI or MDI among either sex (all-P > 0.05). We found that prepregnancy obesity was associated with lower PDI scores at 3 years in boys, but not girls. The mechanisms underlying this sex-specific association remain unclear, but due to elevated obesity exposure in urban populations, further investigation is warranted.
Collapse
Affiliation(s)
- Amy R Nichols
- Department of Nutritional Sciences, University of Texas at Austin, Austin, TX, USA
| | - Andrew G Rundle
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Beverly J Insel
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Lori Hoepner
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Environmental and Occupational Health Sciences, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Virginia Rauh
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Frederica Perera
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Elizabeth M Widen
- Department of Nutritional Sciences, University of Texas at Austin, Austin, TX, USA
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
26
|
Widen EM, Nichols AR, Kahn LG, Factor-Litvak P, Insel BJ, Hoepner L, Dube SM, Rauh V, Perera F, Rundle A. Prepregnancy obesity is associated with cognitive outcomes in boys in a low-income, multiethnic birth cohort. BMC Pediatr 2019; 19:507. [PMID: 31862007 PMCID: PMC6924019 DOI: 10.1186/s12887-019-1853-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/22/2019] [Indexed: 01/13/2023] Open
Abstract
Background Maternal obesity and high gestational weight gain (GWG) disproportionally affect low-income populations and may be associated with child neurodevelopment in a sex-specific manner. We examined sex-specific associations between prepregnancy BMI, GWG, and child neurodevelopment at age 7. Methods Data are from a prospective low-income cohort of African American and Dominican women (n = 368; 44.8% male offspring) enrolled during the second half of pregnancy from 1998 to 2006. Neurodevelopment was measured using the Wechsler Intelligence Scale for Children (WISC-IV) at approximately child age 7. Linear regression estimated associations between prepregnancy BMI, GWG, and child outcomes, adjusting for race/ethnicity, marital status, gestational age at delivery, maternal education, maternal IQ and child age. Results Overweight affected 23.9% of mothers and obesity affected 22.6%. At age 7, full-scale IQ was higher among girls (99.7 ± 11.6) compared to boys (96.9 ± 13.3). Among boys, but not girls, prepregnancy overweight and obesity were associated with lower full-scale IQ scores [overweight β: − 7.1, 95% CI: (− 12.1, − 2.0); obesity β: − 5.7, 95% CI: (− 10.7, − 0.7)]. GWG was not associated with full-scale IQ in either sex. Conclusions Prepregnancy overweight and obesity were associated with lower IQ among boys, but not girls, at 7 years. These findings are important considering overweight and obesity prevalence and the long-term implications of early cognitive development.
Collapse
Affiliation(s)
- Elizabeth M Widen
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, 103 W 24TH ST A2703, Austin, TX, 78712, USA. .,Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.
| | - Amy R Nichols
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, 103 W 24TH ST A2703, Austin, TX, 78712, USA.,Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA
| | - Linda G Kahn
- Department of Pediatrics, New York University School of Medicine, 403 East 34th St, New York, NY, 10016, USA
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA
| | - Beverly J Insel
- Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA.,Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Lori Hoepner
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.,Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA.,Department of Environmental and Occupational Health Sciences, SUNY Downstate Medical Center, School of Public Health, 450 Clarkson Avenue, MSC 43, Brooklyn, NY, 11203, USA
| | - Sara M Dube
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at Austin, 103 W 24TH ST A2703, Austin, TX, 78712, USA.,Department of Nutritional Sciences, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Virginia Rauh
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, 60 Haven Avenue, B-2, Room 213, New York, NY, 10032, USA
| | - Frederica Perera
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA
| | - Andrew Rundle
- Columbia Center for Children's Environmental Health, Mailman School of Public Health, Columbia University, 722 West 168th Street, 12th Floor, New York, NY, 10032, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168 Street Room 1614, New York, NY, 10032, USA
| |
Collapse
|
27
|
Edlow AG, Guedj F, Sverdlov D, Pennings JLA, Bianchi DW. Significant Effects of Maternal Diet During Pregnancy on the Murine Fetal Brain Transcriptome and Offspring Behavior. Front Neurosci 2019; 13:1335. [PMID: 31920502 PMCID: PMC6928003 DOI: 10.3389/fnins.2019.01335] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/27/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Maternal over- and undernutrition in pregnancy plays a critical role in fetal brain development and function. The effects of different maternal diet compositions on intrauterine programing of the fetal brain is a lesser-explored area. The goal of this study was to investigate the impact of two chowmaternal diets on fetal brain gene expression signatures, fetal/neonatal growth, and neonatal and adult behavior in a mouse model. METHODS Throughout pregnancy and lactation, female C57Bl/6J mice were fed one of two standard, commercially available chow diets (pellet versus powder). The powdered chow diet was relatively deficient in micronutrients and enriched for carbohydrates and n-3 long-chain polyunsaturated fatty acids compared to the pelleted chow. RNA was extracted from embryonic day 15.5 forebrains and hybridized to whole genome expression microarrays (N = 5/maternal diet group). Functional analyses of significantly differentially expressed fetal brain genes were performed using Ingenuity Pathways Analysis and Gene Set Enrichment Analysis. Neonatal behavior was assessed using a validated scale (N = 62 pellet-exposed and 31 powder-exposed). Hippocampal learning, locomotor behavior, and motor coordination were assessed in a subset of adults using fear conditioning, open field testing, and Rotarod tests (N = 16 pellet-exposed, 14 powder-exposed). RESULTS Comparing powdered to pelleted chow diets, neither maternal weight trajectory in pregnancy nor embryo size differed. Maternal powdered chow diet was associated with 1647 differentially expressed fetal brain genes. Functional analyses identified significant upregulation of canonical pathways and upstream regulators involved in cell cycle regulation, synaptic plasticity, and sensory nervous system development in the fetal brain, and significant downregulation of pathways related to cell and embryo death. Pathways related to DNA damage response, brain immune response, amino acid and fatty acid transport, and dopaminergic signaling were significantly dysregulated. Powdered chow-exposed neonates were significantly longer but not heavier than pelleted chow-exposed counterparts. On neonatal behavioral testing, powdered chow-exposed neonates achieved coordination- and strength-related milestones significantly earlier, but sensory maturation reflexes significantly later. On adult behavioral testing, powdered chow-exposed offspring exhibited hyperactivity and hippocampal learning deficits. CONCLUSION In wild-type offspring, two diets that differed primarily with respect to micronutrient composition had significant effects on the fetal brain transcriptome, neonatal and adult behavior. These effects did not appear to be mediated by alterations in gross maternal nutritional status nor fetal/neonatal weight. Maternal dietary content is an important variable to consider for investigators evaluating fetal brain development and offspring behavior.
Collapse
Affiliation(s)
- Andrea G. Edlow
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Faycal Guedj
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Deanna Sverdlov
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States
- Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, United States
| | | | - Diana W. Bianchi
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
28
|
Yang S, Mei H, Mei H, Yang Y, Li N, Tan Y, Zhang Y, Zhang D, Zhang Y, Peng A, Zhang B. Risks of maternal prepregnancy overweight/obesity, excessive gestational weight gain, and bottle-feeding in infancy rapid weight gain: evidence from a cohort study in China. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1580-1589. [PMID: 31745693 DOI: 10.1007/s11427-018-9831-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 09/06/2019] [Indexed: 11/30/2022]
Abstract
Rapid weight gain (RWG) in infants is associated with numerous health problems, and its risk factors are still unclear. We assessed 98,097 maternal-infant pairs from a population-based cohort study and followed up with them until the infants were 6 months old. We assessed the associations between maternal prepregnancy weight status; gestational weight gain; feeding pattern; and infants' RWG at 0-1, 0-3, 1-3, and 3-6 months using multivariate unconditional logistic regression models, with controlled confounders. We found that maternal prepregnancy weight status, gestational weight gain, and feeding pattern at the 1st, 3rd, and 6th months had significant impacts on the infants' RWG at each time period (P<0.05). Infants with overweight/obese mothers had a higher risk of RWG after birth, whereas those of mothers who experienced excessive gestational weight gain had higher risks of RWG from birth than the other groups (P<0.01). Infants who were formula-fed had a higher risk of RWG than breastfed infants at the same time point (P<0.01). In conclusion, maternal prepregnancy obesity, excessive gestational weight gain, and formula-feeding were risk factors for infants' RWG during the first 6 months of life.
Collapse
Affiliation(s)
- Shaoping Yang
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Hong Mei
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Hui Mei
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Yan Yang
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Na Li
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Yafei Tan
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Yiming Zhang
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Dan Zhang
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Yan Zhang
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - An'na Peng
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China
| | - Bin Zhang
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, China.
| |
Collapse
|
29
|
Coo H, Fabrigar L, Davies G, Fitzpatrick R, Flavin M. Are observed associations between a high maternal prepregnancy body mass index and offspring IQ likely to be causal? J Epidemiol Community Health 2019; 73:920-928. [PMID: 31358534 DOI: 10.1136/jech-2019-212257] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/18/2019] [Accepted: 07/06/2019] [Indexed: 11/03/2022]
Abstract
BACKGROUND A high maternal prepregnancy body mass index has been associated with lower offspring IQ, but it is unclear if the relationship is causal. To explore this, our objectives were to compare maternal and paternal estimates and to assess whether certain factors mediate the association. METHODS We analysed data from the Avon Longitudinal Study of Parents and Children, which initially recruited 14 541 women residing in Avon, UK, with an expected date of delivery in 1991-1992. Data were collected during and after pregnancy by questionnaire, medical record abstraction and clinical assessment. At approximately 8 years of age, psychologists administered an abbreviated form of the Wechsler Intelligence Scale for Children-III. We fit multivariable logistic regression models to estimate parental prepregnancy obesity and overweight-offspring IQ associations. Counterfactually defined indirect (mediated) effects of maternal prepregnancy obesity on offspring IQ were estimated through path analysis. RESULTS Among 4324 mother-father-child triads and using normal weight as the referent, we observed consistently stronger associations for maternal prepregnancy obesity and offspring performance IQ (eg, adjusted β (95% CI)=-3.4 (-5.7 to -1.2) vs -0.97 (-2.9 to 0.96) for paternal obesity). The indirect effects of maternal obesity on offspring IQ through pathways involving gestational weight gain and duration of breastfeeding were small but significant. CONCLUSION Our findings are consistent with a weak biologic effect of maternal adiposity in pregnancy on offspring performance IQ. Given the growing prevalence of obesity worldwide, more evidence is needed to resolve the correlation versus causation debate in this area.
Collapse
Affiliation(s)
- Helen Coo
- Pediatrics, Queen's University, Kingston, Ontario, Canada
| | | | - Gregory Davies
- Obstetrics & Gynaecology, Queen's University, Kingston, Ontario, Canada
| | | | - Michael Flavin
- Pediatrics, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
30
|
Lei XY, Li YJ, Ou JJ, Li YM. Association between parental body mass index and autism spectrum disorder: a systematic review and meta-analysis. Eur Child Adolesc Psychiatry 2019; 28:933-947. [PMID: 30470952 DOI: 10.1007/s00787-018-1259-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022]
Abstract
Studies have examined the association between parental body mass index (BMI) and autism spectrum disorder (ASD) in offspring, with inconsistent results, especially regarding maternal obesity, overweight and underweight. Cochrane Library, EMBASE, PubMed and PsycINFO databases were searched up to March 2018 for relevant observational studies with no language restriction. Our literature search identified 13 eligible studies for meta-analysis (involving 943,293 children and 30,337 cases). For maternal BMI (13 studies), both maternal obesity [OR 1.41 (95% CI 1.19-1.67)] and maternal overweight [OR 1.16 (95% CI 1.05-1.27)] were significantly associated with ASD, while maternal underweight was not associated with ASD [OR 1.08 (95% CI 0.98-1.20)]. For paternal BMI (three studies), no association was found (paternal obesity: OR 1.28, 95% CI 0.94-1.74; overweight: OR 1.07, 95% CI 0.99-1.15; underweight: OR 1.12, 95% CI 0.87-1.44). Pooled estimates were robust in sensitivity analysis and subgroup analyses. Publication bias may exist for studies assessing maternal BMI and ASD risk, but the filled estimates were not altered. Relative to normal weight, maternal obesity and overweight were significantly associated with increased ASD risk, while maternal underweight was not associated with ASD. Although no association between paternal BMI and ASD was found, current evidence is limited (three studies). Future studies are warranted to address more confounding factors and to identify potential mediators of the association, but pre-pregnancy weight control is suggested.
Collapse
Affiliation(s)
- Xian-Yang Lei
- Office of the President, Central South University, Changsha, 410083, Hunan, China
| | - Yong-Jiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jian-Jun Ou
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ya-Min Li
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
31
|
Risk and Protective Environmental Factors Associated with Autism Spectrum Disorder: Evidence-Based Principles and Recommendations. J Clin Med 2019; 8:jcm8020217. [PMID: 30744008 PMCID: PMC6406684 DOI: 10.3390/jcm8020217] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/03/2019] [Accepted: 02/05/2019] [Indexed: 02/06/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex condition with early childhood onset, characterized by a set of common behavioral features. The etiology of ASD is not yet fully understood; however, it reflects the interaction between genetics and environment. While genetics is now a well-established risk factor, several data support a contribution of the environment as well. This paper summarizes the conclusions of a consensus conference focused on the potential pathogenetic role of environmental factors and on their interactions with genetics. Several environmental factors have been discussed in terms of ASD risk, namely advanced parental age, assisted reproductive technologies, nutritional factors, maternal infections and diseases, environmental chemicals and toxicants, and medications, as well as some other conditions. The analysis focused on their specific impact on three biologically relevant time windows for brain development: the periconception, prenatal, and early postnatal periods. Possible protective factors that might prevent or modify an ASD trajectory have been explored as well. Recommendations for clinicians to reduce ASD risk or its severity have been proposed. Developments in molecular biology and big data approaches, which are able to assess a large number of coexisting factors, are offering new opportunities to disentangle the gene⁻environment interplay that can lead to the development of ASD.
Collapse
|
32
|
Salzwedel AP, Gao W, Andres A, Badger TM, Glasier CM, Ramakrishnaiah RH, Rowell AC, Ou X. Maternal Adiposity Influences Neonatal Brain Functional Connectivity. Front Hum Neurosci 2019; 12:514. [PMID: 30662399 PMCID: PMC6328446 DOI: 10.3389/fnhum.2018.00514] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
The neural mechanisms associated with obesity have been extensively studied, but the impact of maternal obesity on fetal and neonatal brain development remains poorly understood. In this study of full-term neonates, we aimed to detect potential neonatal functional connectivity alterations associated with maternal adiposity, quantified via body-mass-index (BMI) and body-fat-mass (BFM) percentage, based on seed-based and graph theoretical analysis using resting-state fMRI data. Our results revealed significant neonatal functional connectivity alterations in all four functional domains that are implicated in adult obesity: sensory cue processing, reward processing, cognitive control, and motor control. Moreover, some of the detected areas showing regional functional connectivity alterations also showed global degree and efficiency differences. These findings provide important clues to the potential neural basis for cognitive and mental health development in offspring of obese mothers and may lead to the derivation of imaging-based biomarkers for the early identification of risks for timely intervention.
Collapse
Affiliation(s)
- Andrew P Salzwedel
- Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, United States
| | - Wei Gao
- Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, United States.,Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aline Andres
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Thomas M Badger
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Charles M Glasier
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Raghu H Ramakrishnaiah
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Amy C Rowell
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Xiawei Ou
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
33
|
Jung YW, Shim JI, Shim SH, Shin YJ, Shim SH, Chang SW, Cha DH. Global gene expression analysis of cell-free RNA in amniotic fluid from women destined to develop preeclampsia. Medicine (Baltimore) 2019; 98:e13971. [PMID: 30653101 PMCID: PMC6370049 DOI: 10.1097/md.0000000000013971] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Preeclampsia (PE) is a disorder specific to pregnancy characterized by new-onset hypertension and proteinuria after 20 weeks of gestation. There is no definite treatment for PE except delivery of the placenta. The purpose of this study was to elucidate the biological pathways involved in the development of PE and to discover a novel biomarker for PE by performing global gene expression analysis of amniotic fluid cell-free RNA.The participants were recruited from the Department of Obstetrics and Gynecology of CHA Gangnam Medical Center (Seoul, Korea) between March 2014 and February 2015. Eight samples were collected from 8 subjects at second trimester who were later diagnosed with PE. From the amniotic fluid samples, cell-free RNA extraction was performed and gene expression was analyzed using the GeneChip PrimeView Array. Transcriptome data previously analyzed by our group from 9 euploid mid-trimester amniotic fluid samples were used as the control for comparative analysis. Functional analysis of the probe sets was performed using the online Database for Annotation, Visualization, and Integrated Discovery (DAVID) toolkit 6.7.We identified 1841 differentially expressed genes (DEGs) between the PE group and the control. Of these, 1557 genes were upregulated in the PE group, while 284 genes were upregulated in the control. The functional annotation of DEGs identified specific enriched functions such as "transport," "signal transduction," and "stress response." Functional annotation clustering with enriched genes in the PE group revealed that translation-related genes, cell-cell adhesion genes, and immune-related genes were enriched. KEGG pathway analysis showed that several biological pathways, including the ribosome pathway and various immune pathways, were dysregulated. Several genes, including RPS29, IGF-2, and UBC, were significantly upregulated in PE, up to tenfold.This study provides the first genome-wide expression analysis of amniotic fluid cell-free RNA in PE. The results showed that gene expression involving the ribosome pathway and immunologic pathways are dysregulated in PE. Our results will aid in understanding the underlying pathogenesis of PE.
Collapse
Affiliation(s)
- Yong Wook Jung
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
| | - Jung In Shim
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
| | - So Hyun Shim
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
| | - Yun-jeong Shin
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul
| | - Sung Han Shim
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul
| | - Sung Woon Chang
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam-si, Republic of Korea
| | - Dong Hyun Cha
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul
| |
Collapse
|
34
|
Mhatre M, Adeli S, Norwitz E, Craigo S, Phillippe M, Edlow A. The Effect of Maternal Obesity on Placental Cell-Free DNA Release in a Mouse Model. Reprod Sci 2018; 26:1218-1224. [PMID: 30453834 DOI: 10.1177/1933719118811647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The fetal fraction of cell-free DNA (cfDNA) in maternal plasma is decreased in obese women. The underlying mechanism is not well understood. The amount of cfDNA released from the placenta has not been directly examined in maternal obesity. OBJECTIVE We sought to quantify release of cfDNA from the placenta and fetal membranes in maternal diet-induced obesity using explant cultures in an established mouse model. STUDY DESIGN C57BL6/J females were fed either 60% high-fat diet or 10% fat-matched control diet for 14 weeks prepregnancy and throughout gestation. Placentas and fetal membranes were collected on e18 and randomly allocated to time 0-, 1-, or 6-hour culture times. The CfDNA was isolated from culture media, quantified, and normalized to tissue weight. RESULTS Placentas from obese dams released significantly less cfDNA compared to those of lean dams at time 0 (45.8 ± 4.3 ng/mg vs 65.6 ± 7.9 ng/mg, P = .02). Absolute cfDNA levels increased with longer placental culture, with no significant differences between obese and lean dams at 1 and 6 hours. Membranes released significantly less cfDNA than did placentas at every time point. CONCLUSIONS Maternal obesity is associated with decreased release of cfDNA from the placenta compared to lean controls immediately after tissue harvest. This may provide an alternative explanation for the lower fetal fraction of cfDNA noted in maternal obesity.
Collapse
Affiliation(s)
- Mohak Mhatre
- 1 Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| | - Sharareh Adeli
- 2 Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Errol Norwitz
- 1 Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA.,3 Tufts University School of Medicine, Boston, MA, USA
| | - Sabrina Craigo
- 1 Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA.,3 Tufts University School of Medicine, Boston, MA, USA
| | - Mark Phillippe
- 2 Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA.,4 Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA. Edlow is now with Vincent Center for Reproductive Biology and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA, and Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Andrea Edlow
- 1 Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA.,3 Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
35
|
Nteeba J, Ganesan S, Madden JA, Dickson MJ, Keating AF. Progressive obesity alters ovarian insulin, phosphatidylinositol-3 kinase, and chemical metabolism signaling pathways and potentiates ovotoxicity induced by phosphoramide mustard in mice. Biol Reprod 2018; 96:478-490. [PMID: 28203716 DOI: 10.1095/biolreprod.116.143818] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/02/2016] [Accepted: 12/21/2016] [Indexed: 01/01/2023] Open
Abstract
Mechanisms underlying obesity-associated reproductive impairment are ill defined. Hyperinsulinemia is a metabolic perturbation often observed in obese subjects. Insulin activates phosphatidylinositol 3-kinase (PI3K) signaling, which regulates ovarian folliculogenesis, steroidogenesis, and xenobiotic metabolism. The impact of progressive obesity on ovarian genes encoding mRNA involved in insulin-mediated PI3K signaling and xenobiotic biotransformation [insulin receptor (Insr), insulin receptor substrate 1 (Irs1), 2 (Irs2), and 3 (Irs3); kit ligand (Kitlg), stem cell growth factor receptor (Kit), protein kinase B (AKT) alpha (Akt1), beta (Akt2), forkhead transcription factor (FOXO) subfamily 1 (Foxo1), and subfamily 3 (Foxo3a), microsomal epoxide hydrolase (Ephx1), cytochrome P450 family 2, subfamily E, polypeptide 1 (Cyp2e1), glutathione S-transferase (GST) class Pi (Gstp1) and class mu 1 (Gstm1)] was determined in normal wild-type nonagouti (a/a; lean) and lethal yellow mice (KK.CG-Ay/J; obese) at 6, 12, 18, or 24 weeks of age. At 6 weeks, ovaries from obese mice had increased (P < 0.05) Insr and Irs3 but decreased (P < 0.05) Kitlg, Foxo1, and Cyp2e1 mRNA levels. Interestingly, at 12 weeks, an increase (P < 0.05) in Kitlg and Kit mRNA, pIRS1Ser302, pAKTThr308, EPHX1, and GSTP1 protein level was observed due to obesity, while Cyp2e1 mRNA and protein were reduced. A phosphoramide mustard (PM) challenge increased (P < 0.05) ovarian EPHX1 protein abundance in lean but not obese females. In addition, lung tissue from PM-exposed animals had increased (P < 0.05) EPHX1 protein with no impact of obesity thereon. Taken together, progressive obesity affected ovarian signaling pathways potentially involved in obesity-associated reproductive disorders.
Collapse
Affiliation(s)
- Jackson Nteeba
- Department of Animal Science, 2356 Kildee Hall, Iowa State University, Ames, IA, USA
| | - Shanthi Ganesan
- Department of Animal Science, 2356 Kildee Hall, Iowa State University, Ames, IA, USA
| | - Jill A Madden
- Department of Animal Science, 2356 Kildee Hall, Iowa State University, Ames, IA, USA
| | - Mackenzie J Dickson
- Department of Animal Science, 2356 Kildee Hall, Iowa State University, Ames, IA, USA
| | - Aileen F Keating
- Department of Animal Science, 2356 Kildee Hall, Iowa State University, Ames, IA, USA
| |
Collapse
|
36
|
Tarui T, Kim A, Flake A, McClain L, Stratigis JD, Fried I, Newman R, Slonim DK, Bianchi DW. Amniotic fluid transcriptomics reflects novel disease mechanisms in fetuses with myelomeningocele. Am J Obstet Gynecol 2017; 217:587.e1-587.e10. [PMID: 28735706 DOI: 10.1016/j.ajog.2017.07.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Cell-free RNA in amniotic fluid supernatant reflects developmental changes in gene expression in the living fetus, which includes genes that are specific to the central nervous system. Although it has been previously shown that central nervous system-specific transcripts are present in amniotic fluid supernatant, it is not known whether changes in the amniotic fluid supernatant transcriptome reflect the specific pathophysiologic condition of fetal central nervous system disorders. In myelomeningocele, there is open communication between the central nervous system and amniotic fluid. OBJECTIVES The purpose of this study was to identify molecular pathophysiologic changes and novel disease mechanisms that are specific to myelomeningocele by the analysis of amniotic fluid supernatant cell-free RNA in fetuses with open myelomeningocele. STUDY DESIGN Amniotic fluid supernatant was collected from 10 pregnant women at the time of the open myelomeningocele repair in the second trimester (24.5±1.0 weeks); 10 archived amniotic fluid supernatant from sex and gestational age-matched euploid fetuses without myelomeningocele were used as controls (20.9±0.9 weeks). Differentially regulated gene expression patterns were analyzed with the use of human genome expression arrays. RESULTS Fetuses with myelomeningocele had 284 differentially regulated genes (176 up- and 108 down-regulated) in amniotic fluid supernatant. Known genes that were associated with myelomeningocele (PRICKLE2, GLI3, RAB23, HES1, FOLR1) and novel dysregulated genes were identified in association with neurodevelopment and neuronal regeneration (up-regulated, GAP43 and ZEB1) or axonal growth and guidance (down-regulated, ACAP1). Pathway analysis demonstrated a significant contribution of inflammation to disease and a broad influence of Wnt signaling pathways (Wnt1, Wnt5A, ITPR1). CONCLUSION Transcriptomic analyses of living fetuses with myelomeningocele with the use of amniotic fluid supernatant cell-free RNA demonstrated differential regulation of specific genes and molecular pathways relevant to this central nervous system disorder, which resulted in a new understanding of pathophysiologic changes. The data also suggested the importance of pathways that involve secondary disease, such as inflammation, in myelomeningocele. These newly identified pathways may lead to hypotheses that can test novel therapeutic targets as adjuncts to fetal surgical repair.
Collapse
|
37
|
Deardorff J, Smith LH, Petito L, Kim H, Abrams BF. Maternal Prepregnancy Weight and Children's Behavioral and Emotional Outcomes. Am J Prev Med 2017; 53:432-440. [PMID: 28712831 PMCID: PMC5610087 DOI: 10.1016/j.amepre.2017.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 04/13/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022]
Abstract
INTRODUCTION This study investigated associations between maternal prepregnancy BMI and child behaviors at ages 9-11 years and examine interaction by race and gender. METHODS The National Longitudinal Survey of Youth and the Children and Young Adults surveys are U.S.-based, ongoing longitudinal studies, initiated in 1979 and 1986, respectively. Mothers (n=2,952) reported pregnancy and child (n=5,660) developmental information at multiple time points. Child total, internalizing, and externalizing problems at ages 9-11 years were assessed using the Behavior Problems Index (BPI), collected biennially until 2012. Associations between prepregnancy BMI and child BPI outcomes were examined, as well as two- and three-way interactions by race and gender. Analyses were conducted in 2017. RESULTS Boys whose mothers had higher prepregnancy weights exhibited higher total BPI and externalizing scores at ages 9-11 years versus those with normal-weight mothers. Boys with severely obese mothers had higher total BPI (mean difference=7.99, 95% CI=3.53, 12.46) and externalizing (mean difference=5.77, 95% CI=1.50, 10.04) scores. Prepregnancy underweight was associated with boys' higher total BPI (mean difference=2.34, 95% CI=0.02, 4.66) and externalizing (mean difference=3.30, 95% CI=0.69, 5.91); these associations were not significant in sensitivity analyses. No associations emerged for girls or internalizing problems. Two-way interactions by race and three-way interactions by race and gender were not significant. CONCLUSIONS Maternal prepregnancy weight was associated with BPI level among boys. Boys with severely obese mothers exhibited markedly higher behavioral problems at ages 9-11 years versus those with normal-weight mothers, regardless of race. Maintaining healthy prepregnancy weight may be important for preventing boys' deleterious behavior outcomes in middle childhood.
Collapse
Affiliation(s)
- Julianna Deardorff
- Community Health Sciences Division, School of Public Health, University of California, Berkeley, Berkeley, California.
| | - Louisa H Smith
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Lucia Petito
- Division of Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, California
| | - Hyunju Kim
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Barbara F Abrams
- Division of Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, California
| |
Collapse
|
38
|
Jang JH, Jung YW, Shim SH, Sin YJ, Lee KJ, Shim SS, Ahn EH, Cha DH. Global gene expression changes of amniotic fluid cell free RNA according to fetal development. Eur J Obstet Gynecol Reprod Biol 2017; 216:104-110. [DOI: 10.1016/j.ejogrb.2017.07.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/04/2017] [Accepted: 07/10/2017] [Indexed: 11/27/2022]
|
39
|
Widen EM, Kahn LG, Cirillo P, Cohn B, Kezios KL, Factor-Litvak P. Prepregnancy overweight and obesity are associated with impaired child neurodevelopment. MATERNAL AND CHILD NUTRITION 2017. [PMID: 28635158 DOI: 10.1111/mcn.12481] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The authors examined the relationship of prepregnancy body mass index (BMI) and gestational weight gain (GWG) with child neurodevelopment. Mother-child dyads were a subgroup (n = 2,084) of the Child Health and Development Studies from the Oakland, California, area enrolled during pregnancy from 1959 to 1966 and followed at child age 9 years. Linear regression was used to examine associations between prepregnancy BMI, GWG, and standardized Peabody Picture Vocabulary Test and Raven Progressive Matrices scores and to evaluate effect modification of GWG by prepregnancy BMI. Before pregnancy, 77% of women were normal weight, 8% were underweight, 11% were overweight, and 3% were obese. Associations between GWG and child outcomes did not vary by prepregnancy BMI, suggesting no evidence for interaction. In multivariable models, compared to normal prepregnancy BMI, prepregnancy overweight and obesity were associated with lower Peabody scores (b: -1.29; 95% CI [-2.6, -0.04] and b: -2.7; 95% CI [-5.0, -0.32], respectively). GWG was not associated with child Peabody score [b: -0.03 (95% CI: -0.13, 0.07)]. Maternal BMI and GWG were not associated with child Raven score (all P >0.05). Maternal prepregnancy overweight and obesity were associated with lower scores for verbal recognition in mid-childhood. These results contribute to evidence linking maternal BMI with child neurodevelopment. Future research should examine the role of higher prepregnancy BMI values and the pattern of pregnancy weight gain in child cognitive outcomes.
Collapse
Affiliation(s)
- Elizabeth Marie Widen
- Department of Nutritional Sciences, University of Texas at Austin, Austin, Texas, USA.,Department of Medicine, Columbia University Medical Center, New York, New York, USA.,Department of Epidemiology, Columbia University Medical Center, New York, New York, USA
| | - Linda Gross Kahn
- Department of Pediatrics, New York University School of Medicine, New York, New York, USA
| | - Piera Cirillo
- Child Health and Development Studies, Berkeley, California, USA
| | - Barbara Cohn
- Child Health and Development Studies, Berkeley, California, USA
| | - Katrina Lynn Kezios
- Department of Epidemiology, Columbia University Medical Center, New York, New York, USA
| | - Pam Factor-Litvak
- Department of Epidemiology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
40
|
Daraki V, Roumeliotaki T, Koutra K, Georgiou V, Kampouri M, Kyriklaki A, Vafeiadi M, Papavasiliou S, Kogevinas M, Chatzi L. Effect of parental obesity and gestational diabetes on child neuropsychological and behavioral development at 4 years of age: the Rhea mother-child cohort, Crete, Greece. Eur Child Adolesc Psychiatry 2017; 26:703-714. [PMID: 28050707 DOI: 10.1007/s00787-016-0934-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/16/2016] [Indexed: 02/06/2023]
Abstract
Studies have suggested an association between maternal obesity pre-pregnancy and gestational diabetes (GDM) with impaired offspring neurodevelopment, but it is not clear if these associations are explained by shared familiar characteristics. We aimed to assess the associations of maternal and paternal obesity, maternal glucose intolerance in early pregnancy and GDM, with offspring neurodevelopment at 4 years of age. We included 772 mother-child pairs from the "Rhea" Mother-Child cohort in Crete, Greece. Data on maternal/paternal body mass index (BMI) and maternal fasting serum samples for glucose and insulin measurements were collected at 12 weeks of gestation. GDM screening was performed at 24-28 weeks. Neurodevelopment at 4 years was assessed using the McCarthy Scales of Children's Abilities. Behavioral difficulties were assessed by Strengths and Difficulties Questionnaire and Attention Deficit Hyperactivity Disorder Test. Multivariate linear regression analyses showed that maternal obesity was associated with a significant score reduction in general cognitive ability (β-coeff -4.03, 95% CI: -7.08, -0.97), perceptual performance (β-coeff -4.60, 95% CI: -7.74, -1.47), quantitative ability (β-coeff -4.43, 95% CI: -7.68, -1.18), and executive functions (β-coeff -4.92, 95% CI: -8.06, -1.78) at 4 years of age, after adjustment for several confounders and paternal BMI. Maternal obesity was also associated with increased behavioral difficulties (β-coeff 1.22, 95% CI: 0.09, 2.34) and ADHD symptoms (β-coeff 4.28, 95% CI: 1.20, 7.36) at preschool age. Paternal obesity maternal glucose intolerance in early pregnancy and GDM was not associated with child neurodevelopment. These findings suggest that maternal obesity may impair optimal child neurodevelopment at preschool age independently of family shared characteristics.
Collapse
Affiliation(s)
- Vasiliki Daraki
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece.
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Hospital of Crete, Heraklion, Crete, Greece.
| | - Theano Roumeliotaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece
| | - Katerina Koutra
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece
| | - Vaggelis Georgiou
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece
| | - Mariza Kampouri
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece
| | - Andriani Kyriklaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece
| | - Marina Vafeiadi
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece
| | - Stathis Papavasiliou
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Hospital of Crete, Heraklion, Crete, Greece
| | - Manolis Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
| | - Leda Chatzi
- Department of Social Medicine, Faculty of Medicine, University of Crete, PO Box 2208, 71003, Heraklion, Crete, Greece
- Department of Genetics and Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, PO Box 616, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
41
|
Modabbernia A, Velthorst E, Reichenberg A. Environmental risk factors for autism: an evidence-based review of systematic reviews and meta-analyses. Mol Autism 2017; 8:13. [PMID: 28331572 PMCID: PMC5356236 DOI: 10.1186/s13229-017-0121-4] [Citation(s) in RCA: 468] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/12/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND According to recent evidence, up to 40-50% of variance in autism spectrum disorder (ASD) liability might be determined by environmental factors. In the present paper, we conducted a review of systematic reviews and meta-analyses of environmental risk factors for ASD. We assessed each review for quality of evidence and provided a brief overview of putative mechanisms of environmental risk factors for ASD. FINDINGS Current evidence suggests that several environmental factors including vaccination, maternal smoking, thimerosal exposure, and most likely assisted reproductive technologies are unrelated to risk of ASD. On the contrary, advanced parental age is associated with higher risk of ASD. Birth complications that are associated with trauma or ischemia and hypoxia have also shown strong links to ASD, whereas other pregnancy-related factors such as maternal obesity, maternal diabetes, and caesarian section have shown a less strong (but significant) association with risk of ASD. The reviews on nutritional elements have been inconclusive about the detrimental effects of deficiency in folic acid and omega 3, but vitamin D seems to be deficient in patients with ASD. The studies on toxic elements have been largely limited by their design, but there is enough evidence for the association between some heavy metals (most important inorganic mercury and lead) and ASD that warrants further investigation. Mechanisms of the association between environmental factors and ASD are debated but might include non-causative association (including confounding), gene-related effect, oxidative stress, inflammation, hypoxia/ischemia, endocrine disruption, neurotransmitter alterations, and interference with signaling pathways. CONCLUSIONS Compared to genetic studies of ASD, studies of environmental risk factors are in their infancy and have significant methodological limitations. Future studies of ASD risk factors would benefit from a developmental psychopathology approach, prospective design, precise exposure measurement, reliable timing of exposure in relation to critical developmental periods and should take into account the dynamic interplay between gene and environment by using genetically informed designs.
Collapse
Affiliation(s)
- Amirhossein Modabbernia
- Department of Psychiatry and Seaver Autism Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Eva Velthorst
- Department of Psychiatry and Seaver Autism Center, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Abraham Reichenberg
- Department of Psychiatry and Seaver Autism Center, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Friedman Brain Institute, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Seaver Autism Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
42
|
Letra L, Santana I. The Influence of Adipose Tissue on Brain Development, Cognition, and Risk of Neurodegenerative Disorders. ADVANCES IN NEUROBIOLOGY 2017; 19:151-161. [PMID: 28933064 DOI: 10.1007/978-3-319-63260-5_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The brain is a highly metabolic organ and thus especially vulnerable to changes in peripheral metabolism, including those induced by obesity-associated adipose tissue dysfunction. In this context, it is likely that the development and maturation of neurocognitive circuits may also be affected and modulated by metabolic environmental factors, beginning in utero. It is currently recognized that maternal obesity, either pre-gestational or gestational, negatively influences fetal brain development and elevates the risk of cognitive impairment and neuropsychiatric disorders in the offspring. During infancy and adolescence, obesity remains a limiting factor for healthy neurodevelopment, especially affecting executive functions but also attention, visuospatial ability, and motor skills. In middle age, obesity seems to induce an accelerated brain aging and thus may increase the risk of age-related neurodegenerative diseases such as Alzheimer's disease. In this chapter we review and discuss experimental and clinical evidence focusing on the influence of adipose tissue dysfunction on neurodevelopment and cognition across lifespan, as well as some possible mechanistic links, namely the role of the most well studied adipokines.
Collapse
Affiliation(s)
- Liliana Letra
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal. .,Neurology Department, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal.
| | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
43
|
Edlow AG. Maternal obesity and neurodevelopmental and psychiatric disorders in offspring. Prenat Diagn 2016; 37:95-110. [PMID: 27684946 DOI: 10.1002/pd.4932] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 12/19/2022]
Abstract
There is a growing body of evidence from both human epidemiologic and animal studies that prenatal and lactational exposure to maternal obesity and high-fat diet are associated with neurodevelopmental and psychiatric disorders in offspring. These disorders include cognitive impairment, autism spectrum disorders, attention deficit hyperactivity disorder, cerebral palsy, anxiety and depression, schizophrenia, and eating disorders. This review synthesizes human and animal data linking maternal obesity and high-fat diet consumption to abnormal fetal brain development and neurodevelopmental and psychiatric morbidity in offspring. In addition, it highlights key mechanisms by which maternal obesity and maternal diet might impact fetal and offspring neurodevelopment, including neuroinflammation; increased oxidative stress, dysregulated insulin, glucose, and leptin signaling; dysregulated serotonergic and dopaminergic signaling; and perturbations in synaptic plasticity. Finally, the review summarizes available evidence regarding investigational therapeutic approaches to mitigate the harmful effects of maternal obesity on fetal and offspring neurodevelopment. © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Tufts Medical Center, Boston, MA, USA.,Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
44
|
Differences in brain functional connectivity at resting state in neonates born to healthy obese or normal-weight mothers. Int J Obes (Lond) 2016; 40:1931-1934. [PMID: 27677619 DOI: 10.1038/ijo.2016.166] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 08/19/2016] [Accepted: 08/30/2016] [Indexed: 12/31/2022]
Abstract
Recent studies have shown associations between maternal obesity at pre- or early pregnancy and long-term neurodevelopment in children, suggesting in utero effects of maternal obesity on offspring brain development. In this study, we examined whether brain functional connectivity to the prefrontal lobe network is different in newborns from normal-weight or obese mothers. Thirty-four full-term healthy infants from uncomplicated pregnancies were included, with 18 born to normal-weight and 16 born to obese mothers. Two weeks after delivery, the infants underwent an magnetic resonance imaging (MRI) examination during natural sleep, which included structural imaging and resting-state functional MRI (fMRI) scans. Independent component analysis was used to identify the prefrontal lobe network, and dual regression was used to compare functional connectivity between groups. Infants born to normal-weight mothers had higher recruiting (P<0.05, corrected) of dorsal anterior cingulate cortex regions to the prefrontal network after adjusting for maternal intelligence quotient, gestational weight gain and infant postmenstrual age, gender, birth weight/length, head circumference and neonatal diet. The functional connectivity strength in dorsal anterior cingulate cortex negatively correlated (P<0.05) with maternal fat mass percentage measured at early pregnancy. This preliminary study indicates that exposure to maternal obesity in utero may be associated with changes in resting-state functional connectivity in the newborn offspring's brain.
Collapse
|
45
|
Independent and Combined Effects of Maternal Prepregnancy Body Mass Index and Gestational Weight Gain on Offspring Growth at 0-3 Years of Age. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4720785. [PMID: 27652262 PMCID: PMC5019937 DOI: 10.1155/2016/4720785] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/27/2016] [Accepted: 08/02/2016] [Indexed: 02/05/2023]
Abstract
Background. The objective of this study was to investigate the independent and combined effects of maternal prepregnancy body mass index (BMI) and gestational weight gain (GWG) on offspring growth at 0-3 years old. Methods. A total of 826 pairs of nondiabetic mothers and their offspring were recruited in this study. Maternal information was abstracted from medical records and questionnaires. Offspring growth trajectories of weights and BMIs were depicted based on anthropometric measurements. Results. Offspring of mothers who were prepregnancy overweight/obese or obtained excessive GWGs continuously had greater weight and BMI Z-scores throughout the first 3 years of life. Children of prepregnancy overweight/obese mothers with excessive GWGs had a phenotype of higher weight and BMI Z-scores than those prepregnancy overweight/obese ones with nonexcessive GWGs from birth to 18 months. Maternal excessive GWGs increased offspring's risk of overweight/obesity at 12 months (AOR = 1.43, 95% CI: 1.03-2.00) and 24 months (AOR = 1.51, 95% CI: 1.02-2.25). Combination of excessive prepregnancy BMIs and GWGs was significantly associated with offspring's overweight/obesity at 30 months (AOR = 2.98, 95% CI: 1.36-6.53). Conclusions. Maternal prepregnancy overweight/obesity and excessive GWG are both significantly associated with rapid offspring growth from birth to 3 years old. Excessive GWGs strengthen the effects of high maternal prepregnancy BMIs on excessive offspring growth during their early life.
Collapse
|
46
|
Edlow AG, Hui L, Wick HC, Fried I, Bianchi DW. Assessing the fetal effects of maternal obesity via transcriptomic analysis of cord blood: a prospective case-control study. BJOG 2016; 123:180-9. [PMID: 26840378 DOI: 10.1111/1471-0528.13795] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2015] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To analyse fetal gene expression at term using umbilical cord blood, in order to provide insights into the effects of maternal obesity on human development. DESIGN Prospective case-control study. SETTING Academic tertiary care centre. POPULATION Eight obese (body mass index ≥30 kg/m(2)) and eight lean (body mass index <25 kg/m(2)) pregnant women undergoing prelabour caesarean delivery at term. METHODS Women were matched for gestational age and fetal sex. Cord blood RNA was extracted and hybridised to gene expression arrays. Differentially regulated genes were identified using paired t-tests and the Benjamini-Hochberg correction. Functional analyses were performed using Ingenuity Pathway Analysis, BioGPS and Gene Set Enrichment Analysis with a fetal-specific annotation. Z-scores ≥2.0 or P-values <0.01 were considered significant. MAIN OUTCOME MEASURE Functions of differentially regulated genes in fetuses of obese women. RESULTS A total of 701 differentially regulated genes were identified, producing an expression profile implicating neurodegeneration, decreased survival of sensory neurons, and decreased neurogenesis in the fetuses of obese women. Upstream regulators related to inflammatory signalling were significantly activated; those related to insulin receptor signalling, lipid homeostasis, regulation of axonal guidance, and cellular response to oxidative stress were significantly inhibited. Of 26 tissue-specific genes that were differentially regulated in fetuses of obese women, six mapped to the fetal brain. CONCLUSION Maternal obesity affects fetal gene expression at term, implicating dysregulated brain development, inflammatory and immune signalling, glucose and lipid homeostasis, and oxidative stress. This may have implications for postnatal neurodevelopment and metabolism.
Collapse
Affiliation(s)
- A G Edlow
- Mother Infant Research Institute and Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| | - L Hui
- Mother Infant Research Institute and Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA.,Department of Perinatal Medicine, Mercy Hospital for Women, Heidelberg, Vic., Australia
| | - H C Wick
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - I Fried
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - D W Bianchi
- Mother Infant Research Institute and Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
47
|
Mercury as a possible link between maternal obesity and autism spectrum disorder. Med Hypotheses 2016; 91:90-94. [DOI: 10.1016/j.mehy.2016.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/08/2016] [Accepted: 04/13/2016] [Indexed: 01/05/2023]
|
48
|
Edlow AG, Guedj F, Pennings JL, Sverdlov D, Neri C, Bianchi DW. Males are from Mars, and females are from Venus: sex-specific fetal brain gene expression signatures in a mouse model of maternal diet-induced obesity. Am J Obstet Gynecol 2016; 214:623.e1-623.e10. [PMID: 26945603 DOI: 10.1016/j.ajog.2016.02.054] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/05/2016] [Accepted: 02/04/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Maternal obesity is associated with adverse neurodevelopmental outcomes in children, including autism spectrum disorders, developmental delay, and attention-deficit hyperactivity disorder. The underlying mechanisms remain unclear. We previously identified second-trimester amniotic fluid and term cord blood gene expression patterns suggesting dysregulated brain development in fetuses of obese compared with lean women. OBJECTIVE We sought to investigate the biological significance of these findings in a mouse model of maternal diet-induced obesity. We evaluated sex-specific differences in fetal growth, brain gene expression signatures, and associated pathways. STUDY DESIGN Female C57BL/6J mice were fed a 60% high-fat diet or 10% fat control diet for 12-14 weeks prior to mating. During pregnancy, obese dams continued on the high-fat diet or transitioned to the control diet. Lean dams stayed on the control diet. On embryonic day 17.5, embryos were weighed and fetal brains were snap frozen. RNA was extracted from male and female forebrains (10 per diet group per sex) and hybridized to whole-genome expression arrays. Significantly differentially expressed genes were identified using a Welch's t test with the Benjamini-Hochberg correction. Functional analyses were performed using ingenuity pathways analysis and gene set enrichment analysis. RESULTS Embryos of dams on the high-fat diet were significantly smaller than controls, with males more severely affected than females (P = .01). Maternal obesity and maternal obesity with dietary change in pregnancy resulted in significantly more dysregulated genes in male vs female fetal brains (386 vs 66, P < .001). Maternal obesity with and without dietary change in pregnancy was associated with unique brain gene expression signatures for each sex, with an overlap of only 1 gene. Changing obese dams to a control diet in pregnancy resulted in more differentially expressed genes in the fetal brain than maternal obesity alone. Functional analyses identified common dysregulated pathways in both sexes, but maternal obesity and maternal dietary change affected different aspects of brain development in males compared with females. CONCLUSION Maternal obesity is associated with sex-specific differences in fetal size and fetal brain gene expression signatures. Male fetal growth and brain gene expression may be more sensitive to environmental influences during pregnancy. Maternal diet during pregnancy has a significant impact on the embryonic brain transcriptome. It is important to consider both fetal sex and maternal diet when evaluating the effects of maternal obesity on fetal neurodevelopment.
Collapse
|
49
|
Kamath-Rayne BD, Du Y, Hughes M, Wagner EA, Muglia LJ, DeFranco EA, Whitsett JA, Salomonis N, Xu Y. Systems biology evaluation of cell-free amniotic fluid transcriptome of term and preterm infants to detect fetal maturity. BMC Med Genomics 2015; 8:67. [PMID: 26493725 PMCID: PMC4619218 DOI: 10.1186/s12920-015-0138-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/23/2015] [Indexed: 02/03/2023] Open
Abstract
Background Amniotic fluid (AF) is a proximal fluid to the fetus containing higher amounts of cell-free fetal RNA/DNA than maternal serum, thereby making it a promising source for identifying novel biomarkers that predict fetal development and organ maturation. Our aim was to compare AF transcriptomic profiles at different time points in pregnancy to demonstrate unique genetic signatures that would serve as potential biomarkers indicative of fetal maturation. Methods We isolated AF RNA from 16 women at different time points in pregnancy: 4 from 18 to 24 weeks, 6 from 34 to 36 weeks, and 6 from 39 to 40 weeks. RNA-sequencing was performed on cell-free RNA. Gene expression and splicing analyses were performed in conjunction with cell-type and pathway predictions. Results Sample-level analysis at different time points in pregnancy demonstrated a strong correlation with cell types found in the intrauterine environment and fetal respiratory, digestive and external barrier tissues of the fetus, using high-confidence cellular molecular markers. While some RNAs and splice variants were present throughout pregnancy, many transcripts were uniquely expressed at different time points in pregnancy and associated with distinct neonatal co-morbidities (respiratory distress and gavage feeding), indicating fetal immaturity. Conclusion The AF transcriptome exhibits unique cell/organ-selective expression patterns at different time points in pregnancy that can potentially identify fetal organ maturity and predict neonatal morbidity. Developing novel biomarkers indicative of the maturation of multiple organ systems can improve upon our current methods of fetal maturity testing which focus solely on the lung, and will better inform obstetrical decisions regarding delivery timing. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0138-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Beena D Kamath-Rayne
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Yina Du
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Maria Hughes
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Erin A Wagner
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Louis J Muglia
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Emily A DeFranco
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Maternal-Fetal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Nathan Salomonis
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
50
|
Neri C, Edlow AG. Effects of Maternal Obesity on Fetal Programming: Molecular Approaches. Cold Spring Harb Perspect Med 2015; 6:a026591. [PMID: 26337113 DOI: 10.1101/cshperspect.a026591] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Maternal obesity has become a worldwide epidemic. Obesity and a high-fat diet have been shown to have deleterious effects on fetal programming, predisposing offspring to adverse cardiometabolic and neurodevelopmental outcomes. Although large epidemiological studies have shown an association between maternal obesity and adverse outcomes for offspring, the underlying mechanisms remain unclear. Molecular approaches have played a key role in elucidating the mechanistic underpinnings of fetal malprogramming in the setting of maternal obesity. These approaches include, among others, characterization of epigenetic modifications, microRNA expression, the gut microbiome, the transcriptome, and evaluation of specific mRNA expression via quantitative reverse transcription polmerase chain reaction (RT-qPCR) in fetuses and offspring of obese females. This work will review the data from animal models and human fluids/cells regarding the effects of maternal obesity on fetal and offspring neurodevelopment and cardiometabolic outcomes, with a particular focus on molecular approaches.
Collapse
Affiliation(s)
- Caterina Neri
- Department of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome 00100, Italy
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, Massachusetts 02111 Mother Infant Research Institute, Tufts Medical Center, Boston, Massachusetts 02111
| |
Collapse
|