1
|
Zhang H, Lin J, Zhao H. Impacts of Maternal Preeclampsia Exposure on Offspring Neuronal Development: Recent Insights and Interventional Approaches. Int J Mol Sci 2024; 25:11062. [PMID: 39456854 PMCID: PMC11508320 DOI: 10.3390/ijms252011062] [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: 08/28/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Preeclampsia, a hypertensive disorder during pregnancy, frequently correlates with adverse neurological outcomes in offspring, including cognitive impairments, autism spectrum disorder, depressive disorder, attention deficit hyperactivity disorder, and cerebral palsy. Despite these known consequences, the understanding of neuronal damage in the offspring of preeclamptic mothers remains insufficient. Here, we review the neuronal abnormalities resulting from maternal preeclampsia exposure, which include disrupted neurogenesis, loss of neuronal cell integrity, accumulation of cellular debris, decreased synaptogenesis and myelination, and increased neurite growth stimulated by maternal preeclampsia serum. The underlying mechanisms potentially driving these effects involve microglial activation, inflammatory responses, and reduced angiogenesis. Intervention strategies aimed at improving fetal neuronal outcomes are also discussed, encompassing pharmacological treatments such as pravastatin, tadalafil, and melatonin, as well as non-pharmacological approaches like dietary modifications, maternal exercise, and standard care for children. These interventions hold promise for clinical application, offering avenues to address early neuronal abnormalities and prevent the onset of long-term neurological disorders.
Collapse
Affiliation(s)
- He Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Jinju Lin
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| | - Huashan Zhao
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| |
Collapse
|
2
|
Geng L, Qin Z, Han TL, Zhou Y, Zhong X, Zhang G, Dong X. Human amniotic epithelial cells improve uterine spiral artery remodeling to ameliorate preeclampsia in a rat model†. Biol Reprod 2024; 111:906-918. [PMID: 39073898 DOI: 10.1093/biolre/ioae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/26/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024] Open
Abstract
Preeclampsia (PE) is a multisystem pregnancy disorder characterized by impaired remodeling of placental spiral arteries, which leads to the release of pro-inflammatory cytokines and anti-angiogenic agents. However, treatment options for PE are limited, with termination of pregnancy being the only curative option. In this work, we investigated the effects of human amniotic epithelial cells (hAECs) in PE rat model. The rats were induced with lipopolysaccharide (LPS) on gestational day 14.5 followed by injection of hAECs and human umbilical cord mesenchymal stem cells 24 h later. The hAECs treatment resulted in a reduction in blood pressure and proteinuria in the PE rat model. Furthermore, hAECs treatment decreased levels of pro-inflammatory cytokines, reduced inflammatory cells aggregation, and alleviated the damage to placental spiral arteries by downregulating the expression of anti-angiogenic factor and upregulating proangiogenic factor. In vitro experiments confirmed that hAECs treatment restored the proliferation, migration, and angiogenesis of LPS-damaged human umbilical vein endothelial cells. Additionally, hAECs treatment had positive effects on fetal weight and neurological development in the PE group, with no negative effects on the physical development or fertility of offspring rats. These results suggested that hAECs transplantation may be a novel adjuvant therapeutic strategy for PE by reducing the inflammatory and enhancing placental spiral artery angiogenesis.
Collapse
Affiliation(s)
- Lanxin Geng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zuchao Qin
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanqiu Zhou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaocui Zhong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guanghui Zhang
- Chongqing Saiyao Biotechnology Co., Ltd., Chongqing, China
| | - Xiaojing Dong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Tomruk C, Şirin Tomruk C, Denizlioğlu B, Olukman M, Ercan G, Duman S, Köse T, Çetin Uyanıkgil EÖ, Uyanıkgil Y, Uysal A. Effects of apelin on neonatal brain neurogenesis in L-NAME-induced maternal preeclampsia. Sci Rep 2024; 14:19347. [PMID: 39164321 PMCID: PMC11335761 DOI: 10.1038/s41598-024-69326-2] [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: 02/18/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024] Open
Abstract
The aim of this study was to investigate the possible protective effects of apelin, which is known to have antioxidant and anti-inflammatory effects, on changes in neurogenesis in newborns of pregnant rats with L-NAME-induced preeclampsia. Wistar albino female rats were divided into four experimental groups: Control, Apelin, Preeclampsia and Preeclampsia + Apelin. Blood pressure was measured on the 5th, 11th and 17th days of gestation, urine protein was analyzed from urine samples collected for 24 h on the 6th, 12th and 18th days and serum creatinine was analyzed from serum samples. Maternal kidney and placenta tissues were obtained to establish the preeclampsia model, and neonatal brain tissues including the cortex, hippocampus and cerebellum regions were obtained to investigate neurogenesis and examined by histological and immunohistochemical methods. The number of newborns, body weight and brain weight of the newborns were measured. eNOS, IL-10, nNOS and NO levels in the brain analyzed via ELISA. Mean arterial pressure, urine protein and serum creatinine increased in the preeclampsia. Newborn weight decreased in the Preeclampsia group, the values in the Preeclampsia + Apelin group were closer to the Control and Apelin groups. In the Preeclampsia group, edema and dilatation in the proximal and distal tubules of kidneys, perivillous fibrin deposition and increase in syncytial nodules of placenta were observed. VEGF immunoreactivity decreased and iNOS immunoreactivity increased in both kidney and placenta. In neonatal brain tissue examinations, cytotoxic edema accompanied by thinning of cortex, delayed migration and lower cell counts in the hippocampus, and increase in intercellular spaces and EGL thickening in the cerebellum were observed in the preeclampsia. Expression of NeuN, GFAP, MBP, IL-10, eNOS, nNOS and NO levels decreased, whereas expression of Iba-1 increased in the preeclampsia. In the Preeclampsia + Apelin group, these findings were similar to the Control and Apelin groups. Apelin administration was found to be beneficial for preventing the adverse consequences of preeclampsia, but further experimental and clinical studies are needed to better understand these effects.
Collapse
Affiliation(s)
- Canberk Tomruk
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
- Histology and Embryology, Samsun Training and Research Hospital, İlkadim, Samsun, Türkiye
| | - Cansın Şirin Tomruk
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Burcu Denizlioğlu
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
- Emergency Medicine, Aydın State Hospital, Efeler, Aydın, Türkiye
| | - Murat Olukman
- Department of Medical Pharmacology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Gülinnaz Ercan
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Soner Duman
- Department of Internal Medicine, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Timur Köse
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| | - Emel Öykü Çetin Uyanıkgil
- Department of Pharmaceutical Technology, Department of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, Ege University, Bornova, İzmir, Türkiye
| | - Yiğit Uyanıkgil
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye.
| | - Ayşegül Uysal
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Bornova, İzmir, Türkiye
| |
Collapse
|
4
|
Liu H, Wang Z, Li Y, Chen Q, Jiang S, Gao Y, Wang J, Chi Y, Liu J, Wu X, Chen Q, Xiao C, Zhong M, Chen C, Yang X. Hierarchical lncRNA regulatory network in early-onset severe preeclampsia. BMC Biol 2024; 22:159. [PMID: 39075446 PMCID: PMC11287949 DOI: 10.1186/s12915-024-01959-1] [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: 04/12/2023] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Recent studies have shown that several long non-coding RNAs (lncRNAs) in the placenta are associated with preeclampsia (PE). However, the extent to which lncRNAs may contribute to the pathological progression of PE is unclear. RESULTS Here, we report a hierarchical regulatory network involved in early-onset severe PE (EOSPE). We have carried out transcriptome sequencing on the placentae from patients and normal subjects to identify the differentially expressed genes (DEGs), including some lncRNAs (DElncRNAs). We then constructed a high-quality hierarchical regulatory network of lncRNAs, transcription factors (TFs), and target DEGs, containing 1851 lncRNA-TF interactions and 6901 TF-promoter interactions. The lncRNA-to-target regulatory interactions were further validated by the triplex structures between the DElncRNAs and the promoters of the target DEGs. The DElncRNAs in the regulatory network were clustered into 3 clusters, one containing DElncRNAs correlated with the blood pressure, including FLNB-AS1 with targeting 27.89% (869/3116) DEGs in EOSPE. We further demonstrated that FLNB-AS1 could bind the transcription factor JUNB to regulate a series members of the HIF-1 signaling pathway in trophoblast cells. CONCLUSIONS Our results suggest that the differential expression of lncRNAs may perturb the lncRNA-TF-DEG hierarchical regulatory network, leading to the dysregulation of many genes involved in EOSPE. Our study provides a new strategy and a valuable resource for studying the mechanism underlying gene dysregulation in EOSPE patients.
Collapse
Affiliation(s)
- Haihua Liu
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhijian Wang
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yanjun Li
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qian Chen
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sijia Jiang
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yue Gao
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jing Wang
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yali Chi
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jie Liu
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoli Wu
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiong Chen
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoqun Xiao
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mei Zhong
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chunlin Chen
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xinping Yang
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
5
|
Ma Q, Cui Y, Han X, Xiong Y, Xu J, Zhao H, Li X, Cheng W, Zhou Q. Association of maternal hypertension during pregnancy with brain structure and behavioral problems in early adolescence. Eur Child Adolesc Psychiatry 2024; 33:2173-2187. [PMID: 37803213 DOI: 10.1007/s00787-023-02305-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023]
Abstract
Emerging evidence suggests an association between maternal hypertension during pregnancy and mental health in the offspring. However, less is known about the role of hypertensive pregnancy in behavioral symptoms and brain structures of the offspring as well as in their developmental changes. Here, we utilized neuroimaging and behavioral data from 11,878 participants aged 9-10 years and their 2-year follow-up from the Adolescent Brain Cognitive Development (ABCD) study to investigate the long-term effects of maternal hypertension during pregnancy on early adolescent behavior and brain anatomy. Specifically, adolescents born of mothers with maternal hypertension are at risk of long-lasting behavioral problems, as manifested by higher externalizing and internalizing behavior scores at both 9-10 years and 11-12 years. These participants additionally presented with a higher cortical thickness, particularly in the fronto-parieto-temporal areas at 9-10 years. Four regions, including the left parahippocampus, left lateral orbitofrontal lobe, right superior temporal lobe and right temporal pole, remained thicker 2 years later. These findings were partially validated in rats modeled with Nω-nitro-L-arginine methyl ester (L-NAME) preeclampsia. Therefore, clinicians and women who experience hypertension during pregnancy should be warned of this risk, and healthcare providers should recommend appropriate clinical interventions for pregnancy-induced hypertension.
Collapse
Affiliation(s)
- Qing Ma
- Department of Neurology, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China
| | - Yutong Cui
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, 200025, China
| | - Xiaoyang Han
- Department of Neurology, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China
| | - Yu Xiong
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, 200025, China
| | - Jinghui Xu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, 200025, China
| | - Huanqiang Zhao
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, 200025, China
| | - Xiaotian Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China.
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518000, Guangdong, China.
| | - Wei Cheng
- Department of Neurology, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, 321004, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, Shanghai, China.
| | - Qiongjie Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, 200025, China.
| |
Collapse
|
6
|
Check J, Shuster C, Hofheimer J, Camerota M, Dansereau LM, Smith LM, Carter BS, DellaGrotta SA, Helderman J, Kilbride H, Loncar CM, McGowan E, Neal CR, O’Shea TM, Pastyrnak SL, Sheinkopf SJ, Lester BM. Preeclampsia, Fetal Growth Restriction, and 24-Month Neurodevelopment in Very Preterm Infants. JAMA Netw Open 2024; 7:e2420382. [PMID: 38967923 PMCID: PMC11227083 DOI: 10.1001/jamanetworkopen.2024.20382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/04/2024] [Indexed: 07/06/2024] Open
Abstract
Importance Preeclampsia has direct influences on a developing fetus and may impact postnatal health, and fetal growth restriction (FGR) is often seen co-occurring with preeclampsia. The development of children born very preterm after preeclampsia diagnosis with and without FGR is not well characterized. Objective To examine the associations of preeclampsia and FGR with developmental and/or behavioral outcomes in a cohort of very preterm infants. Design, Setting, and Participants In this cohort study, infants in the prospective Neonatal Neurobehavior and Outcomes in Very Preterm Infants study were enrolled between April 2014 and June 2016 from 9 US university-affiliated neonatal intensive care units (NICUs). Eligible infants were born before 30 weeks' gestation. Infants were excluded for any major congenital anomalies and for maternal age younger than 18 years or cognitive impairment impacting the ability to provide informed consent. Data analysis was performed from November 2023 to January 2024. Exposure Maternal preeclampsia and FGR in very preterm infants. Main Outcomes and Measures The Bayley-III cognition, motor, and language scores less than 85 (-1 SD) indicated developmental delay. Child Behavior Checklist/Preschool 1.5-5 T-scores greater than or equal to 64 for internalizing, externalizing, or total problems indicated clinical importance. Results Of 704 infants enrolled, 529 (mean [SD] gestational age, 27.0 [1.9] weeks; 287 male [54.3%]) were studied at 24-month follow-up. A total of 94 infants' mothers had preeclampsia (23.2%), and 46 infants (8.7%) had FGR. In adjusted models, preeclampsia was not associated with Bayley-III (cognitive, B = 3.43 [95% CI, -0.19 to 6.66]; language, B = 3.92 [95% CI, 0.44 to 7.39]; motor, B = 1.86 [95% CI, -1.74 to 5.47]) or Child Behavior Checklist/Preschool 1.5-5 (internalizing, B = -0.08 [95% CI, -2.58 to 2.73]; externalizing, B = 0.69 [95% CI, -1.76 to 3.15]; total, B = 0.21 [95% CI, -2.48 to 2.91]) outcomes. FGR was associated with significantly lower Bayley-III scores (cognitive, B = -8.61 [95% CI, -13.33 to -3.89]; language, B = -8.29 [95% CI, -12.95 to -3.63]; motor, B = -7.60 [95% CI, -12.40 to -2.66]), regardless of preeclampsia status. Conclusions and Relevance In this cohort study of preterm infants, preeclampsia was not associated with developmental and/or behavioral outcomes, but infants with FGR may be prone to developmental delays. These findings suggest future areas of research for understanding the roles of preeclampsia and FGR separately and together in early child development for preterm infants.
Collapse
Affiliation(s)
- Jennifer Check
- Department of Pediatrics, Division of Neonatology, Atrium Health Wake Forest Baptist, Winston-Salem, North Carolina
| | - Coral Shuster
- Department of Pediatrics, Women and Infants Hospital, Providence, Rhode Island
| | - Julie Hofheimer
- Department of Pediatrics, University of North Carolina and Chapel Hill School of Medicine, Chapel Hill
| | - Marie Camerota
- Department of Pediatrics, Women and Infants Hospital, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Lynne M. Dansereau
- Department of Pediatrics, Women and Infants Hospital, Providence, Rhode Island
| | - Lynne M. Smith
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California
| | - Brian S. Carter
- Department of Pediatrics-Neonatology, Children’s Mercy Hospital, Kansas City, Missouri
| | | | - Jennifer Helderman
- Department of Pediatrics, Division of Neonatology, Atrium Health Wake Forest Baptist, Winston-Salem, North Carolina
| | - Howard Kilbride
- Department of Pediatrics-Neonatology, Children’s Mercy Hospital, Kansas City, Missouri
| | - Cynthia M. Loncar
- Department of Pediatrics, Women and Infants Hospital, Providence, Rhode Island
- Department of Pediatrics, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Elisabeth McGowan
- Department of Pediatrics, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Charles R. Neal
- Department of Pediatrics, University of Hawaii John A. Burns School of Medicine, Honolulu
| | - T. Michael O’Shea
- Department of Pediatrics, University of North Carolina and Chapel Hill School of Medicine, Chapel Hill
| | - Steven L. Pastyrnak
- Department of Pediatrics, Spectrum Health-Helen DeVos Hospital, Grand Rapids, Michigan
| | | | - Barry M. Lester
- Department of Pediatrics, Women and Infants Hospital, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island
- Department of Pediatrics, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
7
|
Sun Q, Zhang L, Huang X, Wang M. Salidroside prevents gestational hypertension-induced impairment of offspring learning and memory via Wnt/Skp2 pathway. Neurosci Lett 2024; 832:137787. [PMID: 38641312 DOI: 10.1016/j.neulet.2024.137787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/29/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Salidroside (Sal) has been found to protect against multiple impairments caused by diabetes, and we designed this study to investigate the effect of Sal on gestational hypertension (GHP)-induced impairment of offspring learning and memory. METHODS We established a GHP rat model by intraperitoneal injection of NG-nitro-L-arginine methyl ester (L-NAME), and treated with Sal by daily gavage. We used Morris Water Maze test to evaluate the learning and memory ability of offspring rats. HE staining was used to measured the pathological changes in hippocampus of offspring. Immunohistochemistry, cellular immunofluorescence and western blot were used to detect the protein expression. RESULTS The learning and memory abilities of GHP offspring rats were significantly lower than those of normal rat offspring, while Sal treatment could significantly improve the learning and memory abilities of GHP offspring rats. HE staining did not reveal pathological differences in the hippocampus of normal rats, GHP rats and Sal-treated GHP offspring rats. However, Sal treatment can significantly increase the expression of Wnt1 and Skp2 protein, and decrease the expression of P27kiwf and P21waf1 protein in the hippocampus of GHP offspring rats. In vitro, Sal significantly promoted the proliferation and differentiation on neural stem cell, while Wnt1 knockdown could reverse these promotions by Sal. In the hippocampus of GHP offspring rats, Sal treatment significantly increased the expression of Tuj1, SOX2, Ki67 and DCX protein. CONCLUSION Salidroside significantly improves the learning and memory impairment of offspring caused by GHP, and its mechanism may be related to the fact that Salidroside promotes the proliferation and differentiation of neural stem cells by activating the Wnt1/Skp2 signaling pathway.
Collapse
Affiliation(s)
- Qian Sun
- Department of Gynaecology and Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University Jinan, Shandong 250001, China
| | - Li Zhang
- Department of Gynaecology and Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University Jinan, Shandong 250001, China
| | - Xiuyan Huang
- Department of Gynaecology and Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University Jinan, Shandong 250001, China
| | - Min Wang
- Department of Gynaecology and Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University Jinan, Shandong 250001, China.
| |
Collapse
|
8
|
Griffin A, Bowles T, Solis L, Railey T, Beauti S, Robinson R, Spencer SK, Shaffery JP, Wallace K. Maternal immune suppression during pregnancy does not prevent abnormal behavior in offspring. Biol Sex Differ 2024; 15:27. [PMID: 38532505 DOI: 10.1186/s13293-024-00600-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Offspring of hypertensive disorders of pregnancy are at an increased risk of developing neurodevelopmental and neurobehavioral disorders compared to offspring from non-affected pregnancies. Using rodent models of Preeclampsia (PreE; new onset of hypertension after 20 weeks gestation) and HELLP (hemolysis, elevated liver enzymes, and low platelets), we studied the behavioral outcome of their offspring in adolescence. METHODS A subset of dams received Orencia, a T-cell activation inhibitor, as T cells have been associated with the induction of hypertension and inflammation during pregnancy. We hypothesized that offspring from hypertensive dams would experience adverse behavioral outcomes in social, cognitive, locomotor, and anxiety tests, and offspring from dams treated with Orencia would demonstrate less adverse behaviors. RESULTS Male offspring of PreE + Orencia dams (p < 0.05) and female offspring from HELLP + Orencia dams (p < 0.05) spent more time playing compared to normal pregnant offspring. All offspring from hypertensive and Orencia-treated dams performed worse on the Barnes Maze test compared to normal pregnant. We also measured adult (postnatal day > 60) myelin basic protein (MBP) and NeuN expression in both the prefrontal cortex and hippocampus. In the hippocampus and prefrontal cortex, there was no difference in expression of either MBP or NeuN in all groups regardless of sex. CONCLUSION The results from this study suggest that offspring of hypertensive disorders of pregnancy have behavioral changes, specifically cognitive differences. This study has shown that there is a sex dependent difference in offspring neurobehavioral development, influenced in part by the type of hypertensive disorder of pregnancy, and alterations in the maternal immune system.
Collapse
Affiliation(s)
- Ashley Griffin
- Program in Neuroscience, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Teylor Bowles
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Lucia Solis
- Department of Obstetrics & Gynecology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Teryn Railey
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Samer Beauti
- Master's in Biomedical Science program, School of Graduate Studies in Health Sciences, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Reanna Robinson
- Department of Obstetrics & Gynecology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Shauna-Kay Spencer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - James P Shaffery
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Kedra Wallace
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
- Department of Obstetrics & Gynecology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
9
|
González-Rojas A, Valencia-Narbona M. Neurodevelopmental Disruptions in Children of Preeclamptic Mothers: Pathophysiological Mechanisms and Consequences. Int J Mol Sci 2024; 25:3632. [PMID: 38612445 PMCID: PMC11012011 DOI: 10.3390/ijms25073632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Preeclampsia (PE) is a multisystem disorder characterized by elevated blood pressure in the mother, typically occurring after 20 weeks of gestation and posing risks to both maternal and fetal health. PE causes placental changes that can affect the fetus, particularly neurodevelopment. Its key pathophysiological mechanisms encompass hypoxia, vascular and angiogenic dysregulation, inflammation, neuronal and glial alterations, and disruptions in neuronal signaling. Animal models indicate that PE is correlated with neurodevelopmental alterations and cognitive dysfunctions in offspring and in humans, an association between PE and conditions such as cerebral palsy, autism spectrum disorder, attention deficit hyperactivity disorder, and sexual dimorphism has been observed. Considering the relevance for mothers and children, we conducted a narrative literature review to describe the relationships between the pathophysiological mechanisms behind neurodevelopmental alterations in the offspring of PE mothers, along with their potential consequences. Furthermore, we emphasize aspects pertinent to the prevention/treatment of PE in pregnant mothers and alterations observed in their offspring. The present narrative review offers a current, complete, and exhaustive analysis of (i) the pathophysiological mechanisms that can affect neurodevelopment in the children of PE mothers, (ii) the relationship between PE and neurological alterations in offspring, and (iii) the prevention/treatment of PE.
Collapse
Affiliation(s)
- Andrea González-Rojas
- Laboratorio de Neurociencias Aplicadas, Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Avenida Brasil 2950, Valparaíso 2340025, Chile;
| | | |
Collapse
|
10
|
Troncoso F, Sandoval H, Ibañez B, López-Espíndola D, Bustos F, Tapia JC, Sandaña P, Escudero-Guevara E, Nualart F, Ramírez E, Powers R, Vatish M, Mistry HD, Kurlak LO, Acurio J, Escudero C. Reduced Brain Cortex Angiogenesis in the Offspring of the Preeclampsia-Like Syndrome. Hypertension 2023; 80:2559-2571. [PMID: 37767691 DOI: 10.1161/hypertensionaha.123.21756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Children from pregnancies affected by preeclampsia have an increased risk of cognitive and behavioral alterations via unknown pathophysiology. We tested the hypothesis that preeclampsia generated reduced brain cortex angiogenesis in the offspring. METHODS The preeclampsia-like syndrome (PELS) mouse model was generated by administering the nitric oxide inhibitor NG-nitroarginine methyl ester hydrochloride. Confirmatory experiments were done using 2 additional PELS models. While in vitro analysis used mice and human brain endothelial cells exposed to serum of postnatal day 5 pups or umbilical plasma from preeclamptic pregnancies, respectively. RESULTS We report significant reduction in the area occupied by blood vessels in the motor and somatosensory brain cortex of offspring (postnatal day 5) from PELS compared with uncomplicated control offspring. These data were confirmed using 2 additional PELS models. Furthermore, circulating levels of critical proangiogenic factors, VEGF (vascular endothelial growth factor), and PlGF (placental growth factor) were lower in postnatal day 5 PELS. Also we found lower VEGF receptor 2 (KDR [kinase insert domain-containing receptor]) levels in mice and human endothelial cells exposed to the serum of postnatal day 5 PELS or fetal plasma of preeclamptic pregnancies, respectively. These changes were associated with lower in vitro angiogenic capacity, diminished cell migration, larger F-actin filaments, lower number of filopodia, and lower protein levels of F-actin polymerization regulators in brain endothelial cells exposed to serum or fetal plasma of offspring from preeclampsia. CONCLUSIONS Offspring from preeclampsia exhibited diminished brain cortex angiogenesis, associated with lower circulating VEGF/PlGF/KDR protein levels, impaired brain endothelial migration, and dysfunctional assembly of F-actin filaments. These alterations may predispose to structural and functional alterations in long-term brain development.
Collapse
Affiliation(s)
- Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Hermes Sandoval
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Belén Ibañez
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E., F.B.)
- Group of Research and Innovation in Vascular Health, Chillan, Chile (D.L.-E., C.E.)
| | - Francisca Bustos
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E., F.B.)
| | - Juan Carlos Tapia
- Stem Cells and Neuroscience Center, School of Medicine, University of Talca, Chile (J.C.T.)
| | - Pedro Sandaña
- Anatomopatholy Unit, Hospital Clinico Herminda Martin, Chillan, Chile (P.S.)
| | - Esthefanny Escudero-Guevara
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepcion, Chile (F.N., E.R.)
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile (F.N.)
| | - Eder Ramírez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepcion, Chile (F.N., E.R.)
| | - Robert Powers
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, PA (R.P.)
| | - Manu Vatish
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, England (M.V.)
| | - Hiten D Mistry
- Division of Women and Children's Health, School of Life Course and Population Sciences, King's College London, United Kingdom (H.D.M.)
| | - Lesia O Kurlak
- Stroke Trials Unit, School of Medicine, University of Nottingham, United Kingdom (L.O.K.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
- Group of Research and Innovation in Vascular Health, Chillan, Chile (D.L.-E., C.E.)
| |
Collapse
|
11
|
Liu X, Liu H, Gu N, Pei J, Lin X, Zhao W. Preeclampsia promotes autism in offspring via maternal inflammation and fetal NFκB signaling. Life Sci Alliance 2023; 6:e202301957. [PMID: 37290815 PMCID: PMC10250690 DOI: 10.26508/lsa.202301957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Preeclampsia (PE) is a risk factor for autism spectrum disorder (ASD) in offspring. However, the exact mechanisms underlying the impact of PE on progeny ASD are not fully understood, which hinders the development of effective therapeutic approaches. This study shows the offspring born to a PE mouse model treated by Nω-nitro-L-arginine methyl ester (L-NAME) exhibit ASD-like phenotypes, including neurodevelopment deficiency and behavioral abnormalities. Transcriptomic analysis of the embryonic cortex and adult offspring hippocampus suggested the expression of ASD-related genes was dramatically changed. Furthermore, the level of inflammatory cytokines TNFα in maternal serum and nuclear factor kappa B (NFκB) signaling in the fetal cortex were elevated. Importantly, TNFα neutralization during pregnancy enabled to ameliorate ASD-like phenotypes and restore the NFκB activation level in the offspring exposed to PE. Furthermore, TNFα/NFκB signaling axis, but not L-NAME, caused deficits in neuroprogenitor cell proliferation and synaptic development. These experiments demonstrate that offspring exposed to PE phenocopies ASD signatures reported in humans and indicate therapeutic targeting of TNFα decreases the likelihood of bearing children with ASD phenotypes from PE mothers.
Collapse
Affiliation(s)
- Xueyuan Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
| | - Haiyan Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Nihao Gu
- International Peace Maternity & Child Health Hospital Affiliated to Shanghai Jiao Tong University School of Medicine and Shanghai Key Laboratory for Embryo-Feta Original Adult Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Jiangnan Pei
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xianhua Lin
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Wenlong Zhao
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
- International Peace Maternity & Child Health Hospital Affiliated to Shanghai Jiao Tong University School of Medicine and Shanghai Key Laboratory for Embryo-Feta Original Adult Disease, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
12
|
Nakamura N, Ushida T, Onoda A, Ueda K, Miura R, Suzuki T, Katsuki S, Mizutani H, Yoshida K, Tano S, Iitani Y, Imai K, Hayakawa M, Kajiyama H, Sato Y, Kotani T. Altered offspring neurodevelopment in an L-NAME-induced preeclampsia rat model. Front Pediatr 2023; 11:1168173. [PMID: 37520045 PMCID: PMC10373593 DOI: 10.3389/fped.2023.1168173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction To investigate the mechanism underlying the increased risk of subsequent neurodevelopmental disorders in children born to mothers with preeclampsia, we evaluated the neurodevelopment of offspring of a preeclampsia rat model induced by the administration of N-nitro-L-arginine methyl ester (L-NAME) and identified unique protein signatures in the offspring cerebrospinal fluid. Methods Pregnant rats received an intraperitoneal injection of L-NAME (250 mg/kg/day) during gestational days 15-20 to establish a preeclampsia model. Behavioral experiments (negative geotaxis, open-field, rotarod treadmill, and active avoidance tests), immunohistochemistry [anti-neuronal nuclei (NeuN) staining in the hippocampal dentate gyrus and cerebral cortex on postnatal day 70], and proteome analysis of the cerebrospinal fluid on postnatal day 5 were performed on male offspring. Results Offspring of the preeclampsia dam exhibited increased growth restriction at birth (52.5%), but showed postnatal catch-up growth on postnatal day 14. Several behavioral abnormalities including motor development and vestibular function (negative geotaxis test: p < 0.01) in the neonatal period; motor coordination and learning skills (rotarod treadmill test: p = 0.01); and memory skills (active avoidance test: p < 0.01) in the juvenile period were observed. NeuN-positive cells in preeclampsia rats were significantly reduced in both the hippocampal dentate gyrus and cerebral cortex (p < 0.01, p < 0.01, respectively). Among the 1270 proteins in the cerebrospinal fluid identified using liquid chromatography-tandem mass spectrometry, 32 were differentially expressed. Principal component analysis showed that most cerebrospinal fluid samples achieved clear separation between preeclampsia and control rats. Pathway analysis revealed that differentially expressed proteins were associated with endoplasmic reticulum translocation, Rab proteins, and ribosomal proteins, which are involved in various nervous system disorders including autism spectrum disorders, schizophrenia, and Alzheimer's disease. Conclusion The offspring of the L-NAME-induced preeclampsia model rats exhibited key features of neurodevelopmental abnormalities on behavioral and pathological examinations similar to humans. We found altered cerebrospinal fluid protein profiling in this preeclampsia rat, and the unique protein signatures related to endoplasmic reticulum translocation, Rab proteins, and ribosomal proteins may be associated with subsequent adverse neurodevelopment in the offspring.
Collapse
Affiliation(s)
- Noriyuki Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Anjo Kosei Hospital, Aichi, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Atsuto Onoda
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Kazuto Ueda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Ryosuke Miura
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Toshihiko Suzuki
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Satoru Katsuki
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidesuke Mizutani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukako Iitani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Reproduction and Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
13
|
Escudero C, Kupka E, Ibañez B, Sandoval H, Troncoso F, Wikström AK, López-Espíndola D, Acurio J, Torres-Vergara P, Bergman L. Brain Vascular Dysfunction in Mothers and Their Children Exposed to Preeclampsia. Hypertension 2023; 80:242-256. [PMID: 35920147 DOI: 10.1161/hypertensionaha.122.19408] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Preeclampsia is a maternal syndrome characterized by the new onset of hypertension and proteinuria after 20 weeks of gestation associated with multisystemic complications, including brain alterations. Indeed, brain complications associated with preeclampsia are the leading direct causes of fetal and maternal morbidity and mortality, especially in low- and middle-income countries. In addition to the well-recognized long-term adverse cardiovascular effects of preeclampsia, women who have had preeclampsia have higher risk of stroke, dementia, intracerebral white matter lesions, epilepsy, and perhaps also cognitive decline postpartum. Furthermore, increasing evidence has also associated preeclampsia with similar cognitive and cerebral disorders in the offspring. However, the mechanistic links between these associations remain unresolved. This article summarizes the current knowledge about the cerebrovascular complications elicited by preeclampsia and the potential pathophysiological mechanisms involved, emphasizing the impaired brain vascular function in the mother and their offspring.
Collapse
Affiliation(s)
- Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.).,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.)
| | - Ellen Kupka
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Sweden (E.K.)
| | - Belen Ibañez
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Hermes Sandoval
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Sweden (A.K.W., L.B.)
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.).,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.)
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.).,Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Chile (P.T.-V.)
| | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Sweden (A.K.W., L.B.).,Department of Obstetrics and Gynecology, Stellenbosch University, South Africa (L.B.).,Department of clinical sciences, Sahlgrenska Academy, Gothenburg University, Sweden (L.B.)
| |
Collapse
|
14
|
Barron A, Manna S, McElwain CJ, Musumeci A, McCarthy FP, O’Keeffe GW, McCarthy CM. Maternal pre-eclampsia serum increases neurite growth and mitochondrial function through a potential IL-6-dependent mechanism in differentiated SH-SY5Y cells. Front Physiol 2023; 13:1043481. [PMID: 36714304 PMCID: PMC9877349 DOI: 10.3389/fphys.2022.1043481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction: Pre-eclampsia (PE) is a common and serious hypertensive disorder of pregnancy, which affects 3%-5% of first-time pregnancies and is a leading cause of maternal and neonatal morbidity and mortality. Prenatal exposure to PE is associated with an increased risk of neurodevelopmental disorders in affected offspring, although the cellular and molecular basis of this increased risk is largely unknown. Methods: Here, we examined the effects of exposure to maternal serum from women with PE or a healthy uncomplicated pregnancy on the survival, neurite growth and mitochondrial function of neuronally differentiated human SH-SY5Y neuroblastoma cells, which are commonly used to study neurite growth. Neurite growth and mitochondrial function are two strongly linked neurodevelopmental parameters in which alterations have been implicated in neurodevelopmental disorders. Following this, we investigated the pleiotropic cytokine interleukin-6 (IL-6) levels as a potential mechanism. Results: Cells exposed to 3% (v/v) PE serum for 72 h exhibited increased neurite growth (p < 0.05), which was validated in the human neural progenitor cell line, ReNcell® VM (p < 0.01), and mitochondrial respiration (elevated oxygen consumption rate (p < 0.05), basal mitochondrial respiration, proton leak, ATP synthesis, and non-mitochondrial respiration) compared to control serum-treated cells. ELISA analysis showed elevations in maternal IL-6 in PE sera (p < 0.05) and placental explants (p < 0.05). In support of this, SH-SY5Y cells exposed to 3% (v/v) PE serum for 24 h had increased phospho-STAT3 levels, which is a key intracellular mediator of IL-6 signalling (p < 0.05). Furthermore, treatment with anti-IL-6 neutralizing antibody blocked the effects of PE serum on neurite growth (p < 0.05), and exposure to IL-6 promoted neurite growth in SH-SY5Y cells (p < 0.01). Discussion: Collectively these data show elevated serum levels of maternal IL-6 in PE, which increases neurite growth and mitochondrial function in SH-SY5Y cells. This rationalizes the further study of IL-6 as a potential mediator between PE exposure and neurodevelopmental outcome in the offspring.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Samprikta Manna
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Colm J. McElwain
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Andrea Musumeci
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Gerard W. O’Keeffe
- Department of Anatomy and Neuroscience, University College, Cork, Ireland,Cork Neuroscience Centre, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland,*Correspondence: Gerard W. O’Keeffe, ; Cathal M. McCarthy,
| |
Collapse
|
15
|
Environmental Enrichment Protects Offspring of a Rat Model of Preeclampsia from Cognitive Decline. Cell Mol Neurobiol 2023; 43:381-394. [PMID: 35119541 DOI: 10.1007/s10571-022-01192-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/07/2022] [Indexed: 01/07/2023]
Abstract
Preeclampsia affects 5-7% of all pregnancies and contributes to adverse pregnancy and birth outcomes. In addition to the short-term effects of preeclampsia, preeclampsia can exert long-term adverse effects on offspring. Numerous studies have demonstrated that offspring of preeclamptic women exhibit cognitive deficits from childhood to old age. However, effective ways to improve the cognitive abilities of these offspring remain to be investigated. The aim of this study was to explore whether environmental enrichment in early life could restore the cognitive ability of the offspring of a rat model of preeclampsia and to investigate the cellular and molecular mechanisms by which EE improves cognitive ability. L-NAME was used to establish a rat model of preeclampsia. The spatial learning and memory abilities and recognition memory of 56-day-old offspring were evaluated by the Morris water maze and Novel object recognition (NOR) task. Immunofluorescence was performed to evaluate cell proliferation and apoptosis in the DG region of the hippocampus. qRT-PCR was performed to examine the expression levels of neurogenesis-associated genes, pre- and postsynaptic proteins and inflammatory cytokines. An enzyme-linked immune absorbent assay was performed to evaluate the concentration of vascular endothelial growth factor (VEGF) and inflammatory cytokines in the hippocampus. The administration of L-NAME led to increased systolic blood pressure and urine protein levels in pregnant rats. Offspring in the L-NAME group exhibited impaired spatial learning ability and memory as well as NOR memory. Hippocampal neurogenesis and synaptic plasticity were impaired in offspring from the L-NAME group. Furthermore, cell apoptosis in the hippocampus was increased in the L-NAME group. The hippocampus was skewed to a proinflammatory profile, as shown by increased inflammatory cytokine levels. EE improved the cognitive ability of offspring in the L-NAME group and resulted in increased hippocampal neurogenesis and synaptic protein expression levels and decreased apoptosis and inflammatory cytokine levels. Environmental enrichment resolves cognitive impairment in the offspring of a rat model of preeclampsia by improving hippocampal neurogenesis and synaptic plasticity and normalizing the apoptosis level and the inflammatory balance.
Collapse
|
16
|
Torres Y, Celis C, Acurio J, Escudero C. Language Impairment in Children of Mothers with Gestational Diabetes, Preeclampsia, and Preterm Delivery: Current Hypothesis and Potential Underlying Mechanisms : Language Impartment and Pregnancy Complications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:245-267. [PMID: 37466777 DOI: 10.1007/978-3-031-32554-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Many conditions may impair or delay language development, including socioeconomic status, parent's education, or intrauterine environment. Accordingly, increasing evidence has described that pregnancy complications, including gestational diabetes mellitus (GDM), preeclampsia, and preterm delivery, are associated with the offspring's impaired neurodevelopment. Since language is one of the high brain functions, alterations in this function are another sign of neurodevelopment impairment. How these maternal conditions may generate language impairment has yet to be entirely understood. However, since language development requires adequate structural formation and function/connectivity of the brain, these processes must be affected by alterations in maternal conditions. However, the underlying mechanisms of these structural alterations are largely unknown. This manuscript critically analyzes the literature focused on the risk of developing language impairment in children of mothers with GDM, preeclampsia, and preterm delivery. Furthermore, we highlight potential underlying molecular mechanisms associated with these alterations, such as neuroinflammatory and metabolic and cerebrovascular alterations.
Collapse
Affiliation(s)
- Yesenia Torres
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile
- Brainlab-Cognitive Neuroscience Research Group, Department of Clinical Psychology and Psychobiology, University of Barcelona, Barcelona, Catalonia, Spain
| | - Cristian Celis
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile
- Centro terapéutico , ABCfonoaudiologia, Santiago, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile.
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.
| |
Collapse
|
17
|
Ma Y, Yang Y, Lv M, Zhang Y, He Q, Zhang Y, Su H, Deng X, Qian Y. 1,25(OH) 2D 3 alleviates LPS-induced preeclampsia-like rats impairment in the protective effect by TLR4/NF-kB pathway. Placenta 2022; 130:34-41. [PMID: 36372042 DOI: 10.1016/j.placenta.2022.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Accumulating epidemiological studies support that Vitamin D deficiency is associated with the pathogenesis of preeclampsia. However, it is unknown whether vitamin D can be used as a treatment for preeclampsia. This study aimed to explore whether vitamin D supplementation could improve the rat model of preeclampsia. METHODS LPS was used to establish a rat model of preeclampsia. Inflammatory cytokines were examined by QRT-PCR and ELISA assays, and the concentration of sfit-1 and NO was assessed by ELISA. Analyzing the pathological features of the placenta with hematoxylin-eosin. The spatial learning and memory abilities of offspring were evaluated by the Morris water maze. Immune histology and western blot were performed to evaluate the expression levels of inflammatory pathway-associated Factor and vascular endothelium-associated Factor in the placenta. RESULTS Vitamin D treatment reduced the blood pressure and urine protein of PE model rats, alleviated pathological damage to the placenta and pregnancy outcomes, and protected PE offspring from impaired memory and learning abilities. Moreover, TLR4 signaling pathway in the placenta was inhibited. Furthermore, vitamin D supplementation increased the expression of endothelial growth factor and vascular relaxing factor, and there was no significant difference compared with the control group. DISCUSSION We generated the result that Vitamin D supplementation significantly improved the phenotype of preeclampsia and adverse pregnancy outcome caused by an abnormal inflammatory reaction and endothelial dysfunction in the placenta, and improved the learning and cognitive ability of offspring.
Collapse
Affiliation(s)
- Yantuanjin Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Yuling Yang
- Department of Cell Biology and Medical Genetics, Kunming Medical University, Kunming, China
| | - Mengxin Lv
- Department of Medical Genetics and Prenatal Diagnosis, Kunming Maternal and Child Health Hospital Kunming, China
| | - Yuhang Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Qiuyue He
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Yaqin Zhang
- Department of Reproduction, The First People's Hospital of Puer City, Puer, China
| | - Hong Su
- Department of Reproduction, The First People's Hospital of Puer City, Puer, China
| | - Xingli Deng
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Yuan Qian
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Laboratory Medicine, Kunming, China; Department of Cell Biology and Medical Genetics, Kunming Medical University, Kunming, China; Department of Medical Genetics and Prenatal Diagnosis, Kunming Maternal and Child Health Hospital Kunming, China.
| |
Collapse
|
18
|
de Alwis N, Binder NK, Beard S, Mangwiro YTM, Kadife E, Cuffe JSM, Keenan E, Fato BR, Kaitu’u-Lino TJ, Brownfoot FC, Marshall SA, Hannan NJ. The L-NAME mouse model of preeclampsia and impact to long-term maternal cardiovascular health. Life Sci Alliance 2022; 5:5/12/e202201517. [PMID: 36260752 PMCID: PMC9356384 DOI: 10.26508/lsa.202201517] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia affects ∼2–8% of pregnancies worldwide. It is associated with increased long-term maternal cardiovascular disease risk. This study assesses the effect of the vasoconstrictor N(ω)-nitro-L-arginine methyl ester (L-NAME) in modelling preeclampsia in mice, and its long-term effects on maternal cardiovascular health. In this study, we found that L-NAME administration mimicked key characteristics of preeclampsia, including elevated blood pressure, impaired fetal and placental growth, and increased circulating endothelin-1 (vasoconstrictor), soluble fms-like tyrosine kinase-1 (anti-angiogenic factor), and C-reactive protein (inflammatory marker). Post-delivery, mice that received L-NAME in pregnancy recovered, with no discernible changes in measured cardiovascular indices at 1-, 2-, and 4-wk post-delivery, compared with matched controls. At 10-wk post-delivery, arteries collected from the L-NAME mice constricted significantly more to phenylephrine than controls. In addition, these mice had increased kidney Mmp9:Timp1 and heart Tnf mRNA expression, indicating increased inflammation. These findings suggest that though administration of L-NAME in mice certainly models key characteristics of preeclampsia during pregnancy, it does not appear to model the adverse increase in cardiovascular disease risk seen in individuals after preeclampsia.
Collapse
Affiliation(s)
- Natasha de Alwis
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
- Mercy Perinatal, Heidelberg, Australia
| | - Natalie K Binder
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
- Mercy Perinatal, Heidelberg, Australia
| | - Sally Beard
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
- Mercy Perinatal, Heidelberg, Australia
| | - Yeukai TM Mangwiro
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
- Mercy Perinatal, Heidelberg, Australia
| | - Elif Kadife
- Mercy Perinatal, Heidelberg, Australia
- Department of Obstetrics and Gynaecology, Obstetrics Diagnostics and Therapeutics Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
| | - James SM Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Emerson Keenan
- Mercy Perinatal, Heidelberg, Australia
- Department of Obstetrics and Gynaecology, Obstetrics Diagnostics and Therapeutics Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
| | - Bianca R Fato
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
- Mercy Perinatal, Heidelberg, Australia
| | - Tu’uhevaha J Kaitu’u-Lino
- Mercy Perinatal, Heidelberg, Australia
- Department of Obstetrics and Gynaecology, Diagnostics Discovery and Reverse Translation in Pregnancy Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
| | - Fiona C Brownfoot
- Mercy Perinatal, Heidelberg, Australia
- Department of Obstetrics and Gynaecology, Obstetrics Diagnostics and Therapeutics Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, The Ritchie Centre, School of Clinical Sciences, Monash University and The Hudson Institute of Medical Research, Clayton, Australia
| | - Natalie J Hannan
- Department of Obstetrics and Gynaecology, Therapeutics Discovery and Vascular Function Group, The University of Melbourne and Mercy Hospital for Women, Heidelberg, Australia
- Mercy Perinatal, Heidelberg, Australia
| |
Collapse
|
19
|
Lara E, Rivera N, González-Bernal A, Rojas D, López-Espíndola D, Rodríguez A, Escudero C. Abnormal cerebral microvascular perfusion and reactivity in female offspring of reduced uterine perfusion pressure (RUPP) mice model. J Cereb Blood Flow Metab 2022; 42:2318-2332. [PMID: 36008921 PMCID: PMC9670000 DOI: 10.1177/0271678x221121872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 12/14/2022]
Abstract
Children born from women with preeclampsia have alterations in cerebral neurovascular development and a high risk for developing cognitive alterations. Because cerebral blood vessels are critical components in cerebrovascular development, we evaluated the brain microvascular perfusion and microvascular reactivity (exposed to external stimuli of warm and cold) in pups born to preeclampsia-like syndrome based on the reduction of uterine perfusion (RUPP). Also, we evaluate the angiogenic proteomic profile in those brains. Pregnant mice showed a reduction in uterine flow after RUPP surgery (-40 to 50%) associated with unfavorable perinatal results compared to sham mice. Furthermore, offspring of the RUPP mice exhibited reduced brain microvascular perfusion at postnatal day 5 (P5) compared with offspring from sham mice. This reduction was preferentially observed in females. Also, brain microvascular reactivity to external stimuli (warm and cold) was reduced in pups of RUPP mice. Furthermore, a differential expression of the angiogenic profile associated with inflammation, extrinsic apoptotic, cancer, and cellular senescence processes as the primary signaling impaired process was found in the brains of RUPP-offspring. Then, offspring (P5) from preeclampsia-like syndrome exhibit impaired brain perfusion and microvascular reactivity, particularly in female mice, associated with differential expression of angiogenic proteins in the brain tissue.
Collapse
Affiliation(s)
- Evelyn Lara
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
| | - Nathaly Rivera
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
| | - Alejandro González-Bernal
- Department of Clinical Sciences, Faculty of Veterinary Sciences,
Universidad de Concepción, Chillán, Chile
| | - Daniela Rojas
- Department of Pathology, Faculty of Veterinary Sciences,
Universidad de Concepción, Chillán, Chile
| | - Daniela López-Espíndola
- School of Medical Technology and Biomedical Research Center,
Faculty of Medicine, Universidad de Valparaíso
- Group of Research and Innovation in Vascular Health (GRIVAS
Health), Chillán, Chile
| | - Andrés Rodríguez
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS
Health), Chillán, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS
Health), Chillán, Chile
| |
Collapse
|
20
|
Cumsille P, Lara E, Verdugo-Hernández P, Acurio J, Escudero C. A robust quantitative approach for laser speckle contrast imaging perfusion analysis revealed anomalies in the brain blood flow in offspring mice of preeclampsia. Microvasc Res 2022; 144:104418. [PMID: 35931124 DOI: 10.1016/j.mvr.2022.104418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 02/01/2023]
Abstract
Microcirculation analysis of the brain cortex is challenging because surface perfusion varies rapidly in small space-time regions and is bone protected. The laser speckle contrast imaging (LSCI) technique allows analyzing in vivo brain vascular perfusion generating a large amount of data that requires sophisticated data analytics, making researchers invest much effort in processing. Our research question was whether the reduced placental perfusion model (RUPP) of preeclampsia (PE) was associated with impaired blood perfusion in the offspring's brains. We aimed to develop a robust numerical approach that mainly consisted of applying a signal-processing tool for calculating optimal segmentation and piece-wise fits of the offspring's brain perfusion signals obtained from the LSCI technique. We combined this tool with the usual statistical analysis, implementing both in Matlab software. We performed brain perfusion measurements from offspring (five days postnatal, P5) of control pregnant dams (sham, n = 13) and of RUPP dams (RUPP, n = 7) using the Pericam® PSI-HR system at a basal condition and after thermal stimuli (warm and cold). We found that pups of RUPP mice exhibited significant differences in perfusion and vascular response to thermal stimuli compared to the sham mice. These differences were associated with high data variability in the Sham group, while in the RUPP group, perfusion looks "stiffer." Data also suggest sex-dimorphism in the vascular response since female pups in the Sham group but not male pups showed statistically significant differences in response to the warm stimulus. Again, this sex-related difference was absent in pups of RUPP mice. In conclusion, we present a robust quantitative approach for LSCI measurements that revealed anomalies in the brain blood flow in offspring of the RUPP model of PE.
Collapse
Affiliation(s)
- Patricio Cumsille
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Centre for Biotechnology and Bioengineering (CeBiB), University of Chile, Santiago, Chile.
| | - Evelyn Lara
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Paula Verdugo-Hernández
- Escuela de Pedagogía en Ciencias Naturales y Exactas, Facultad de Ciencias de la Educación, Universidad de Talca, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.
| |
Collapse
|
21
|
Placental dysfunction: The core mechanism for poor neurodevelopmental outcomes in the offspring of preeclampsia pregnancies. Placenta 2022; 126:224-232. [PMID: 35872512 DOI: 10.1016/j.placenta.2022.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022]
Abstract
Preeclampsia (PE) is a leading condition threatening pregnant women and their offspring. The offspring of PE pregnancies have a high risk of poor neurodevelopmental outcomes and neuropsychological diseases later in life. However, the pathophysiology and pathogenesis of poor neurodevelopment remain undetermined. Abnormal placental functions are at the core of most PE cases, and recent research evidence supports that the placenta plays an important role in fetal brain development. Here, we summarize the relationship between abnormal fetal brain development and placental dysfunction in PE conditions, which include the dysfunction of nutrient and gas-waste exchange, impaired angiogenesis stimulation, abnormal neurotransmitter regulation, disrupted special protectors, and immune disorders. All these factors could lead to poor neurodevelopmental outcomes.
Collapse
|
22
|
Systemic Maternal Human sFLT1 Overexpression Leads to an Impaired Foetal Brain Development of Growth-Restricted Foetuses upon Experimental Preeclampsia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3024032. [PMID: 35693702 PMCID: PMC9184195 DOI: 10.1155/2022/3024032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/19/2022] [Indexed: 11/29/2022]
Abstract
The pregnancy disorder preeclampsia (PE) is characterized by maternal hypertension, increased level of circulating antiangiogenic soluble fms-like tyrosine kinase-1 (sFLT1), and reduced placental perfusion, leading to foetal growth restriction (FGR) and preterm birth. All these adverse effects are associated with neurocognitive disorders in the offspring. However, the direct interplay between increased antiangiogenesis during PE and disturbed foetal brain development independent of prematurity has not been investigated yet. To examine foetal brain development in sFLT1-related PE, hsFLT1/rtTA-transgenic mice with systemic (maternal or maternal/fetoplacental) human sFLT1 (hsFLT1) overexpression since 10.5 days postconception (dpc) were used, and histological and molecular analyses of foetal brains were performed at 18.5 dpc. Consequences of elevated hsFLT1 on placental/foetal vascularization and hypoxia of placentas and foetal brains were analysed using the hypoxia markers pimonidazole and hemeoxygenase-1 (HO-1). Immunohistochemical analysis revealed increased hypoxia in placentas of PE-affected pregnancies. Moreover, an increase in HO-1 expression was observed upon elevated hsFLT1 in placentas and foetal brains. PE foetuses revealed asymmetrical FGR by increased brain/liver weight ratio. The brain volume was reduced combined with a reduction in the cortical/hippocampal area and an increase of the caudate putamen and its neuroepithelium, which was associated with a reduced cell density in the cortex and increased cell density in the caudate putamen upon hsFLT1 overexpression. Mild influences were observed on brain vasculature shown by free iron deposits and mRNA changes in Vegf signalling. Of note, both types of systemic hsFLT1 overexpression (indirect: maternal or direct: maternal/fetoplacental) revealed similar changes with increasing severity of impaired foetal brain development. Overall, circulating hsFLT1 in PE pregnancies impaired uteroplacental perfusion leading to disturbed foetal oxygenation and brain injury. This might be associated with a disturbed cell migration from the caudate putamen neuroepithelium to the cortex which could be due to disturbed cerebrovascular adaption.
Collapse
|
23
|
Selivanova EK, Shvetsova AA, Borzykh AA, Gaynullina DK, Kiryukhina OO, Lukoshkova EV, Potekhina VM, Kuzmin VS, Tarasova OS. Intrauterine L-NAME Exposure Weakens the Development of Sympathetic Innervation and Induces the Remodeling of Arterial Vessels in Two-Week-Old Rats. Int J Mol Sci 2021; 22:ijms222212327. [PMID: 34830206 PMCID: PMC8618620 DOI: 10.3390/ijms222212327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 11/19/2022] Open
Abstract
Nitric oxide (NO) has been shown to stimulate differentiation and increase the survival of ganglionic sympathetic neurons. The proportion of neuronal NOS-immunoreactive sympathetic preganglionic neurons is particularly high in newborn rats and decreases with maturation. However, the role of NO in the development of vascular sympathetic innervation has never been studied before. We tested the hypothesis that intrauterine NO deficiency weakened the development of vascular sympathetic innervation and thereby changed the contractility of peripheral arteries and blood pressure level in two-week-old offspring. Pregnant rats consumed NOS inhibitor L-NAME (250 mg/L in drinking water) from gestational day 10 until delivery. Pups in the L-NAME group had a reduced body weight and blood level of NO metabolites at 1–2 postnatal days. Saphenous arteries from two-week-old L-NAME offspring demonstrated a lower density of sympathetic innervation, a smaller inner diameter, reduced maximal active force and decreased α-actin/β-actin mRNA expression ratio compared to the controls. Importantly, pups in the L-NAME group exhibited decreased blood pressure levels before, but not after, ganglionic blockade with chlorisondamine. In conclusion, intrauterine L-NAME exposure is followed by the impaired development of the sympathetic nervous system in early postnatal life, which is accompanied by the structural and functional remodeling of arterial blood vessels.
Collapse
Affiliation(s)
- Ekaterina K. Selivanova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (E.K.S.); (A.A.S.); (D.K.G.); (V.M.P.); (V.S.K.)
| | - Anastasia A. Shvetsova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (E.K.S.); (A.A.S.); (D.K.G.); (V.M.P.); (V.S.K.)
| | - Anna A. Borzykh
- Laboratory of Exercise Physiology, State Research Center of the Russian Federation, Institute for Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia;
| | - Dina K. Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (E.K.S.); (A.A.S.); (D.K.G.); (V.M.P.); (V.S.K.)
| | - Oxana O. Kiryukhina
- Laboratory for the Study of Information Processes at the Cellular and Molecular Levels, Institute for Information Transmission Problems, Russian Academy of Sciences, 119333 Moscow, Russia;
| | - Elena V. Lukoshkova
- Laboratory of Experimental Pathology of the Heart, National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia;
| | - Viktoria M. Potekhina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (E.K.S.); (A.A.S.); (D.K.G.); (V.M.P.); (V.S.K.)
| | - Vladislav S. Kuzmin
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (E.K.S.); (A.A.S.); (D.K.G.); (V.M.P.); (V.S.K.)
| | - Olga S. Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (E.K.S.); (A.A.S.); (D.K.G.); (V.M.P.); (V.S.K.)
- Laboratory of Exercise Physiology, State Research Center of the Russian Federation, Institute for Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia;
- Correspondence:
| |
Collapse
|
24
|
Ushida T, Kidokoro H, Nakamura N, Katsuki S, Imai K, Nakano-Kobayashi T, Moriyama Y, Sato Y, Hayakawa M, Natsume J, Kajiyama H, Kotani T. Impact of maternal hypertensive disorders of pregnancy on brain volumes at term-equivalent age in preterm infants: A voxel-based morphometry study. Pregnancy Hypertens 2021; 25:143-149. [PMID: 34139669 DOI: 10.1016/j.preghy.2021.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Infants born to mothers with hypertensive disorders of pregnancy (HDP) reportedly have negative behavioral and neurodevelopmental outcomes. However, the effects of maternal HDP on infant brain growth have not been fully evaluated. We aimed to evaluate brain volumes and brain injury in preterm infants born to mothers with HDP using magnetic resonance (MR) imaging at term-equivalent age. STUDY DESIGN In this cohort study, MR imaging was performed for 94 preterm infants born before 34 weeks of gestation at Nagoya University Hospital between 2010 and 2018. Twenty infants were born to mothers with HDP and 74 to mothers without HDP. MAIN OUTCOME MEASURES Total brain volumes and regional volumetric alterations were assessed by voxel-based morphometry, and brain injury was evaluated using the Kidokoro global brain abnormality score. Developmental quotient was assessed at a corrected age of 1.5 years in 59 infants (HDP, n = 11; non-HDP, n = 48). RESULTS No significant differences were observed in the gray and white matter volumes of the two groups (HDP: 175 ± 24 mL, 137 ± 13 mL, respectively; non-HDP: 172 ± 24 mL, 142 ± 13 mL, respectively). Additionally, no regional volumetric alterations were observed between the two groups after covariate adjustment (gestational age and infant sex). The total Kidokoro score and developmental quotient were similar in both groups. CONCLUSIONS No significant differences in the global and regional brain volumes were observed. Further research is needed to confirm our findings at different time points of MR imaging and in different populations.
Collapse
Affiliation(s)
- Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoru Katsuki
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Nakano-Kobayashi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshinori Moriyama
- Department of Obstetrics and Gynecology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Jun Natsume
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Developmental Disability Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Perinatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
25
|
The novel testicular enrichment protein Cfap58 is required for Notch-associated ciliogenesis. Biosci Rep 2021; 40:221781. [PMID: 31904090 PMCID: PMC6970087 DOI: 10.1042/bsr20192666] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/28/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Cilia and flagella are critical organelles with conserved internal structures and diverse developmental and physiological processes according to cell type. Although the core components of structures are shared with thousands of associated proteins involved in cilia or flagella formation, we hypothesized that some unknown proteins, such as outer dense fiber 2 (Odf2/Cenexin) perform distinct functions in these organelles. In the present study, we identified several uncharacterized proteins through mass spectrometry interactome analysis of Odf2/Cenexin proteins. We further examined the expression patterns and functions of a protein named cilia and flagella associated protein 58 (Cfap58) in cultured astrocytes and sperm flagella. The results of a combination of biochemical analyses and drug administration studies reveal that Cfap58 is a testis-enrichment protein that exhibits similar localization to Odf2/Cenexin proteins and is required for the elongation of the primary cilium and sperm midpiece via modulation of the Notch signaling pathway. However, the cell cycle-related functions and localization of Odf2/Cenexin in the mother centriole were not altered in Cfap58 knockdown cells. These findings indicate that Cfap58 may be partially recruited by Odf2/Cenexin proteins and is indispensable for the cilia and flagellar assembly. These data provide us with a better understanding of ciliogenesis and flagellar elongation and may aid in identifying new targets for diseases caused by Notch-mediated ciliopathies and flagellar abnormalities.
Collapse
|
26
|
Ren Z, Gao Y, Gao Y, Liang G, Chen Q, Jiang S, Yang X, Fan C, Wang H, Wang J, Shi YW, Xiao C, Zhong M, Yang X. Distinct placental molecular processes associated with early-onset and late-onset preeclampsia. Am J Cancer Res 2021; 11:5028-5044. [PMID: 33754042 PMCID: PMC7978310 DOI: 10.7150/thno.56141] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Patients with preeclampsia display a spectrum of onset time and severity of clinical presentation, yet the underlying molecular bases for the early-onset and late-onset clinical subtypes are not known. Although several transcriptome studies have been done on placentae from PE patients, only a small number of differentially expressed genes have been identified due to very small sample sizes and no distinguishing of clinical subtypes. Methods: We carried out RNA-seq on 65 high-quality placenta samples, including 33 from 30 patients and 32 from 30 control subjects, to search for dysregulated genes and the molecular network and pathways they are involved in. Results: We identified two functionally distinct sets of dysregulated genes in the two major subtypes: 2,977 differentially expressed genes in early-onset severe preeclampsia, which are enriched with metabolism-related pathways, notably transporter functions; and 375 differentially expressed genes in late-onset severe preeclampsia, which are enriched with immune-related pathways. We also identified some key transcription factors, which may drive the widespread gene dysregulation in both early-onset and late-onset patients. Conclusion: These results suggest that early-onset and late-onset severe preeclampsia have different molecular mechanisms, whereas the late-onset mild preeclampsia may have no placenta-specific causal factors. A few regulators may be the key drivers of the dysregulated molecular pathways.
Collapse
|
27
|
Gumusoglu SB, Chilukuri ASS, Hing BWQ, Scroggins SM, Kundu S, Sandgren JA, Santillan MK, Santillan DA, Grobe JL, Stevens HE. Altered offspring neurodevelopment in an arginine vasopressin preeclampsia model. Transl Psychiatry 2021; 11:79. [PMID: 33510137 PMCID: PMC7844013 DOI: 10.1038/s41398-021-01205-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/17/2020] [Accepted: 01/08/2021] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia is a severe gestational hypertensive condition linked to child neuropsychiatric disorders, although underlying mechanisms are unclear. We used a recently developed, clinically relevant animal model of preeclampsia to assess offspring. C57BL/6J mouse dams were chronically infused with arginine vasopressin (AVP) or saline (24 ng/h) throughout pregnancy. Adult offspring were behaviorally tested (Y-maze, open field, rotarod, social approach, and elevated plus maze). Offspring brain was assessed histologically and by RNA sequencing. Preeclampsia-exposed adult males exhibited increased anxiety-like behavior and social approach while adult females exhibited impaired procedural learning. Adult AVP-exposed males had reduced total neocortical volume. Adult AVP-exposed females had increased caudate-putamen volume, increased caudate-putamen cell number, and decreased excitatory synapse density in hippocampal dentate gyrus (DG), CA1, and CA3. At postnatal day 7 (P7), AVP-exposed male and female offspring both had smaller neocortex. At P7, AVP-exposed males also had smaller caudate-putamen volume, while females had increased caudate-putamen volume relative to neocortical size. Similar to P7, E18 AVP-exposed offspring had smaller dorsal forebrain, mainly in reduced intermediate, subventricular, and ventricular zone volume, particularly in males. Decreased volume was not accounted for by cell size or cerebrovascular vessel diameter changes. E18 cortical RNAseq revealed 49 differentially-expressed genes in male AVP-exposed offspring, over-representing cytoplasmic translation processes. In females, 31 genes were differentially-expressed, over-representing collagen-related and epithelial regulation pathways. Gene expression changes in E18 AVP-exposed placenta indicated potential underlying mechanisms. Deficits in behavior and forebrain development in this AVP-based preeclampsia model were distinctly different in males and females, implicating different neurobiological bases.
Collapse
Affiliation(s)
- Serena Banu Gumusoglu
- Interdisciplinary Neuroscience Graduate Program, University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
| | - Akanksha Sri Satya Chilukuri
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Benjamin Wen Qing Hing
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sabrina Marie Scroggins
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sreelekha Kundu
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Jeremy Anton Sandgren
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mark Kharim Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Donna Ann Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Justin Lewis Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hanna Elizabeth Stevens
- Interdisciplinary Neuroscience Graduate Program, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
28
|
Hypertensive disorders of pregnancy and the risk of offspring depression in childhood: Findings from the Avon Longitudinal Study of Parents and Children. Dev Psychopathol 2020; 32:845-851. [PMID: 31345273 DOI: 10.1017/s0954579419000944] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Hypertensive disorders of pregnancy (HDP) may increase the risk of offspring depression in childhood. Low birth weight is also associated with increased risk of mental health problems, including depression. This study sought to investigate (a) whether there is an association between HDP and the risk of depression in childhood and (b) whether low birth weight mediates this association. The current study is based on the Avon Longitudinal Study of Parents and Children (ALSPAC), a prospective, population-based study that has followed a cohort of offspring since their mothers were pregnant (n = 6,739). Depression at the age of 7 years was diagnosed using parent reports via the Development and Well-Being Assessment (DAWBA). Log-binomial regression and mediation analyses were used. Children exposed to HDP were 2.3 times more likely to have a depression diagnosis compared with nonexposed children, adjusted Risk Ratio [RR], 2.31; 95% CI, [1.20, 4.47]. Low birth weight was a weak mediator of this association. Results were adjusted for confounding variables including antenatal depression and anxiety during pregnancy.This study suggests that fetal exposure to maternal hypertensive disorders of pregnancy increased the risk of childhood depression. The study adds to the evidence suggesting that the uterine environment is a critical determinant of neurodevelopmental and psychiatric outcomes.
Collapse
|
29
|
Preeclampsia-Associated lncRNA INHBA-AS1 Regulates the Proliferation, Invasion, and Migration of Placental Trophoblast Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 22:684-695. [PMID: 33230466 PMCID: PMC7585871 DOI: 10.1016/j.omtn.2020.09.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023]
Abstract
Preeclampsia is believed to be caused by impaired placentation with insufficient trophoblast invasion, leading to impaired uterine spiral artery remodeling and angiogenesis. However, the underlying molecular mechanism remains unknown. We recently carried out transcriptome profiling of placental long noncoding RNAs (lncRNAs) and identified 383 differentially expressed lncRNAs in early-onset severe preeclampsia. Here, we are reporting our identification of lncRNA INHBA-AS1 as a potential causal factor of preeclampsia and its downstream pathways that may be involved in placentation. We found that INHBA-AS1 was upregulated in patients and positively correlated with clinical severity. We systematically searched for potential INHBA-AS1-binding transcription factors and their targets in databases and found that the targets were enriched with differentially expressed genes in the placentae of patients. We further demonstrated that the lncRNA INHBA-AS1 inhibited the invasion and migration of trophoblast cells through restraining the transcription factor CENPB from binding to the promoter of TNF receptor-associated factor 1 (TRAF1). Therefore, we have identified the dysregulated pathway "INHBA-AS1-CENPB-TRAF1" as a contributor to the pathogenesis of preeclampsia through prohibiting the proliferation, invasion, and migration of trophoblasts during placentation.
Collapse
|
30
|
Kay VR, Rätsep MT, Figueiró-Filho EA, Croy BA. Preeclampsia may influence offspring neuroanatomy and cognitive function: a role for placental growth factor†. Biol Reprod 2020; 101:271-283. [PMID: 31175349 DOI: 10.1093/biolre/ioz095] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023] Open
Abstract
Preeclampsia (PE) is a common pregnancy complication affecting 3-5% of women. Preeclampsia is diagnosed clinically as new-onset hypertension with associated end organ damage after 20 weeks of gestation. Despite being diagnosed as a maternal syndrome, fetal experience of PE is a developmental insult with lifelong cognitive consequences. These cognitive alterations are associated with distorted neuroanatomy and cerebrovasculature, including a higher risk of stroke. The pathophysiology of a PE pregnancy is complex, with many factors potentially able to affect fetal development. Deficient pro-angiogenic factor expression is one aspect that may impair fetal vascularization, alter brain structure, and affect future cognition. Of the pro-angiogenic growth factors, placental growth factor (PGF) is strongly linked to PE. Concentrations of PGF are inappropriately low in maternal blood both before and during a PE gestation. Fetal concentrations of PGF appear to mirror maternal circulating concentrations. Using Pgf-/- mice that may model effects of PE on offspring, we demonstrated altered central nervous system vascularization, neuroanatomy, and behavior. Overall, we propose that development of the fetal brain is impaired in PE, making the offspring of preeclamptic pregnancies a unique cohort with greater risk of altered cognition and cerebrovasculature. These individuals may benefit from early interventions, either pharmacological or environmental. The early neonatal period may be a promising window for intervention while the developing brain retains plasticity.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Matthew T Rätsep
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
31
|
Marcelle ET, Oliva MT, Hinshaw SP. Gestational Smoking and Hypertension as Predictors of Working Memory Functioning in Childhood Attention-Deficit/Hyperactivity Disorder. Front Psychol 2020; 11:1950. [PMID: 33041878 PMCID: PMC7518085 DOI: 10.3389/fpsyg.2020.01950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/14/2020] [Indexed: 01/03/2023] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental condition affecting between 5 and 8% of all children and adolescents, characterized by impairing levels of inattention, hyperactivity, and impulsivity. Existing cognitive models of ADHD have placed working memory (WM) deficits at the core of ADHD and suggest that primary WM deficits may also underlie the additional deficits associated with the condition. Although not all children with ADHD show WM deficits, those with such deficits have been found to have worse functional outcomes when compared to their diagnosed peers with typical WM functioning. Even so, contributors to the variability of impaired WM functioning seen within this population remain unknown. In the present study, we examined the association between two known prenatal and perinatal risk factors for impaired cognitive functioning - gestational smoking and hypertension - in three independent samples of children and adolescents with ADHD (samples varied with respect to sample size and WM measurement procedures). Contrary to hypotheses and existing literature, presence of high blood pressure during pregnancy was unexpectedly found to be a positive predictor of offspring WM capacity in one of three samples (a sample of all girls with ADHD). Implications and considerations for future research are discussed.
Collapse
Affiliation(s)
- Enitan T. Marcelle
- Department of Psychology, University of California, Berkeley, Berkeley, CA, United States
| | - Mercedes T. Oliva
- Department of Psychology, University of California, Berkeley, Berkeley, CA, United States
| | - Stephen P. Hinshaw
- Department of Psychology, University of California, Berkeley, Berkeley, CA, United States
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Gumusoglu SB, Chilukuri ASS, Santillan DA, Santillan MK, Stevens HE. Neurodevelopmental Outcomes of Prenatal Preeclampsia Exposure. Trends Neurosci 2020; 43:253-268. [PMID: 32209456 DOI: 10.1016/j.tins.2020.02.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/21/2020] [Accepted: 02/05/2020] [Indexed: 01/06/2023]
Abstract
Preeclampsia is a dangerous hypertensive disorder of pregnancy with known links to negative child health outcomes. Here, we review epidemiological and basic neuroscience work from the past several decades linking prenatal preeclampsia to altered neurodevelopment. This work demonstrates increased rates of neuropsychiatric disorders [e.g., increased autism spectrum disorder, attention deficit hyperactivity disorder (ADHD)] in children of preeclamptic pregnancies, as well as increased rates of cognitive impairments [e.g., decreased intelligence quotient (IQ), academic performance] and neurological disease (e.g., stroke and epilepsy). We also review findings from multiple animal models of preeclampsia. Manipulation of key clinical preeclampsia processes in these models (e.g., placental hypoxia, immune dysfunction, angiogenesis, oxidative stress) causes various disruptions in offspring, including ones in white matter/glia, glucocorticoid receptors, neuroimmune outcomes, cerebrovascular structure, and cognition/behavior. This animal work implicates potentially high-yield targets that may be leveraged in the future for clinical application.
Collapse
Affiliation(s)
- Serena B Gumusoglu
- Department of Psychiatry, University of Iowa Carver College of Medicine, Department of Psychiatry, Iowa City, IA, USA
| | - Akanksha S S Chilukuri
- Department of Psychiatry, University of Iowa Carver College of Medicine, Department of Psychiatry, Iowa City, IA, USA
| | - Donna A Santillan
- University of Iowa Carver College of Medicine, Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - Mark K Santillan
- University of Iowa Carver College of Medicine, Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Department of Psychiatry, Iowa City, IA, USA.
| |
Collapse
|
33
|
Ijomone OK, Shallie PD, Naicker T. Oligodendrocytes Death Induced Sensorimotor and Cognitive Deficit in N-nitro-L-arginine methyl Rat Model of Pre-eclampsia. Neurochem Res 2020; 45:902-914. [PMID: 31983010 DOI: 10.1007/s11064-020-02969-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/17/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
Abstract
Pre-eclampsia (PE) is a pregnancy complicated syndrome that affects multiple organs including the brain that continue post- delivery in both mother and the offspring. We evaluated the expression of oligodendrocytes in the brain of PE rat model through development as well as the cognitive changes and other behavioural modifications that may occur later in the life of offspring of PE-like rat model. Pregnant rats divided into early-onset and late-onset groups were administered with N-nitro- L-arginine methyl (L-NAME) through drinking water at gestational days (GD) 8-17. Rats were allowed free access to water throughout the pregnancy. At GD 19, post-natal day (PND) 1 and 60, rats were sacrificed and brain excised for further analysis. The offspring were subjected to behavioural studies for cognitive and sensorimotor impairments before sacrificed at PND 60. Results showed significant down-regulation in the expression of OLIG2 in PE at GD 19 brain which persists till PND 60. Likewise, there was a significant increase in the latency to locate the platform in Morris water maze, time to traverse the balance beam and reduced hanging time on the wire test between the control and the PE treated. PE could lead to impaired neuronal signalling through demyelination which may contributes significantly to long-term sensorimotor and cognitive deficit.
Collapse
Affiliation(s)
- Olayemi K Ijomone
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Philemon Dauda Shallie
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
34
|
Nꭃ-nitro-l-arginine methyl model of pre-eclampsia elicits differential IBA1 and EAAT1 expressions in brain. J Chem Neuroanat 2019; 100:101660. [DOI: 10.1016/j.jchemneu.2019.101660] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/21/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
|
35
|
Lu HQ, Hu R. Lasting Effects of Intrauterine Exposure to Preeclampsia on Offspring and the Underlying Mechanism. AJP Rep 2019; 9:e275-e291. [PMID: 31511798 PMCID: PMC6736667 DOI: 10.1055/s-0039-1695004] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/12/2019] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia is a common pregnancy complication which can have adverse impact on both mother and baby. In addition to the short term effects, a large body of epidemiological evidence has found preeclampsia can exert long-lasting effects on mother and offspring. Studies suggest that offspring exposed to preeclampsia are at a higher risk of developing cardiovascular, metabolic, and neurological diseases, as well as other diseases. However, studies investigating the underlying mechanism are limited, the exact mechanism still remains unclear. In this study, we will review the epidemiological evidence and studies exploring the mechanism underlying long-term effects of preeclampsia on offspring. Further studies should be targeted at this field so as to implement effective clinical management to prevent the exposed offspring from potential diseases.
Collapse
Affiliation(s)
- Hui Qing Lu
- Department of Obstetrics, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Rong Hu
- Department of Obstetrics, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Lara E, Acurio J, Leon J, Penny J, Torres-Vergara P, Escudero C. Are the Cognitive Alterations Present in Children Born From Preeclamptic Pregnancies the Result of Impaired Angiogenesis? Focus on the Potential Role of the VEGF Family. Front Physiol 2018; 9:1591. [PMID: 30487752 PMCID: PMC6246680 DOI: 10.3389/fphys.2018.01591] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022] Open
Abstract
Evidence from clinical studies has proposed that children born from preeclamptic women have a higher risk of suffering neurological, psychological, or behavioral alterations. However, to date, the mechanisms behind these outcomes are poorly understood. Here, we speculate that the neurodevelopmental alterations in the children of preeclamptic pregnancies result from impaired angiogenesis. The pro-angiogenic factors vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) are key regulators of both vascular and neurological development, and it has been widely demonstrated that umbilical blood of preeclamptic pregnancies contains high levels of soluble VEGF receptor type 1 (sFlt-1), a decoy receptor of VEGF. As a consequence, this anti-angiogenic state could lead to long-lasting neurological outcomes. In this non-systematic review, we propose that alterations in the circulating concentrations of VEGF, PlGF, and sFlt-1 in preeclamptic pregnancies will affect both fetal cerebrovascular function and neurodevelopment, which in turn may cause cognitive alterations in post-natal life.
Collapse
Affiliation(s)
- Evelyn Lara
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - José Leon
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Department of Pharmacy, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (LFV-GIANT), Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Red Iberoamericana de alteraciones Vasculares Asociadas a TRastornos del EMbarazo (RIVA-TREM), Chillán, Chile
| |
Collapse
|
37
|
Kay VR, Rätsep MT, Cahill LS, Hickman AF, Zavan B, Newport ME, Ellegood J, Laliberte CL, Reynolds JN, Carmeliet P, Tayade C, Sled JG, Croy BA. Effects of placental growth factor deficiency on behavior, neuroanatomy, and cerebrovasculature of mice. Physiol Genomics 2018; 50:862-875. [PMID: 30118404 DOI: 10.1152/physiolgenomics.00076.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Preeclampsia, a hypertensive syndrome occurring in 3-5% of human pregnancies, has lifelong health consequences for fetuses. Cognitive ability throughout life is altered, and adult stroke risk is increased. One potential etiological factor for altered brain development is low concentrations of proangiogenic placental growth factor (PGF). Impaired PGF production may promote an antiangiogenic fetal environment during neural and cerebrovascular development. We previously reported delayed vascularization of the hindbrain, altered retinal vascular organization, and less connectivity in the circle of Willis in Pgf-/- mice. We hypothesized Pgf-/- mice would have impaired cognition and altered brain neuroanatomy in addition to compromised cerebrovasculature. Cognitive behavior was assessed in adult Pgf-/- and Pgf+/+ mice by four paradigms followed by postmortem high-resolution MRI of neuroanatomy. X-ray microcomputed tomography imaging investigated the three-dimensional cerebrovascular geometry in another cohort. Pgf-/- mice exhibited poorer spatial memory, less depressive-like behavior, and superior recognition of novel objects. Significantly smaller volumes of 10 structures were detected in the Pgf-/- compared with Pgf+/+ brain. Pgf-/- brain had more total blood vessel segments in the small-diameter range. Lack of PGF altered cognitive functions, brain neuroanatomy, and cerebrovasculature in mice. Pgf-/- mice may be a preclinical model for the offspring effects of low-PGF preeclampsia gestation.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Bruno Zavan
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada.,Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais , Brazil
| | - Margaret E Newport
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | | | - James N Reynolds
- Centre for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB - Vesalius Research Center, University of Leuven, Department of Oncology , Leuven , Belgium
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada.,Department of Medical Biophysics, University of Toronto , Ontario , Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
38
|
Zhang Z, Wang X, Wang J, Zhang L. The decreased expression of Stat3 and p-Stat3 in preeclampsia-like rat placenta. J Mol Histol 2018; 49:175-183. [DOI: 10.1007/s10735-018-9757-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/18/2018] [Indexed: 11/29/2022]
|
39
|
Zhao W, Li Z, Ping P, Wang G, Yuan X, Sun F. Outer dense fibers stabilize the axoneme to maintain sperm motility. J Cell Mol Med 2017; 22:1755-1768. [PMID: 29168316 PMCID: PMC5824370 DOI: 10.1111/jcmm.13457] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/13/2017] [Indexed: 11/28/2022] Open
Abstract
Outer dense fibers (ODFs), as unique accessory structures in mammalian sperm, are considered to play a role in the protection of the sperm tail against shear forces. However, the role and relevant mechanisms of ODFs in modulating sperm motility and its pathological involvement in asthenozoospermia were unknown. Here, we found that the percentage of ODF defects was higher in asthenozoospermic samples than that in control samples and was significantly correlated with the percentage of axoneme defects and non-motile sperm. Furthermore, the expression levels of ODF major components (Odf1, 2, 3, 4) were frequently down-regulated in asthenozoospermic samples. Intriguingly, the positive relationship between ODF size and sperm motility existed across species. The conditional disruption of Odf2 expression in mice led to reduced sperm motility and the characteristics of asthenozoospermia. Meanwhile, the expression of acetylated α-tubulin was decreased in sperm from both Odf2 conditional knockout (cKO) mice and asthenozoospermic men. Immunofluorescence and biochemistry analyses showed that Odf2 could bind to acetylated α-tubulin and protect the acetylation level of α-tubulin in HEK293T cells in a cold environment. Finally, we found that lithium elevated the expression levels of Odf family proteins and acetylated α-tubulin, elongated the midpiece length and increased the percentage of rapidly moving sperm in mice. Our results demonstrate that ODFs are beneficial for sperm motility via stabilization of the axoneme and that hypo-expression of Odf family proteins is involved in the pathogenesis of asthenozoospermia. The lithium administration assay will provide valuable insights into the development of new treatments for asthenozoospermia.
Collapse
Affiliation(s)
- Wenlong Zhao
- International Peace Maternity & Child Health Hospital, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhengzheng Li
- International Peace Maternity & Child Health Hospital, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ping Ping
- International Peace Maternity & Child Health Hospital, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Department of Assistant Reproduction, International Peace Maternity & Child Health Hospital, Shanghai, China
| | - Guishuan Wang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Xiaobing Yuan
- Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China.,Hussman Institute for Autism, Baltimore, MD, USA
| | - Fei Sun
- International Peace Maternity & Child Health Hospital, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
40
|
Zhu H, Zhu W, Hu R, Wang H, Ma D, Li X. The effect of pre-eclampsia-like syndrome induced by L-NAME on learning and memory and hippocampal glucocorticoid receptor expression: A rat model. Hypertens Pregnancy 2016; 36:36-43. [PMID: 27786581 DOI: 10.1080/10641955.2016.1228957] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE We aimed to study the impacts of pre-eclampsia on the cognitive and learning capabilities of adolescent rat offspring and to explore the possible underlying mechanisms at the molecular level. METHODS Pregnant rats were subcutaneously injected with saline solution (control) (n = 16) or NG-nitro-L-arginine methyl ester (L-NAME) (n = 16) from the 13th day of gestation until parturition. The brain tissues from fetal rats delivered by cesarean section were examined in both groups with hematoxylin and eosin (H&E) staining. Rats born vaginally in both groups were subjected to the Morris water maze test when 8-week-old and their hippocampi were analyzed for glucocorticoid receptor (GR) expression. RESULTS A pre-eclampsia-like model was successfully built in pregnant rats by infusion of the NO synthase inhibitor L-NAME, including phenotypes as maternal hypertension and proteinuria, high stillbirth rate, and fetal growth retardation. Neuroepithelial cell proliferation was found in the hippocampus of fetal rats in the L-NAME group. Grown to 8-week-old, the L-NAME group showed significantly longer escape latency than the control group in the beginning as well as in the end of navigation trials. At the same time, the swimming distance achieved by the L-NAME group was significantly longer than that of the control group. Such differences in cognitive and learning capabilities between the two groups were not gender dependent. Besides, the 8-week-old rats in the L-NAME group had increased GR expression in the hippocampus than the control group. CONCLUSION Pre-eclampsia would impair cognitive and learning capabilities in adolescent offspring, and the upregulated expression of hippocampal GR may be involved in the underlying mechanisms.
Collapse
Affiliation(s)
- Hao Zhu
- a Obstetrics & Gynecology Hospital , Shanghai , China
| | - Weimin Zhu
- a Obstetrics & Gynecology Hospital , Shanghai , China
| | - Rong Hu
- a Obstetrics & Gynecology Hospital , Shanghai , China
| | - Huijun Wang
- b Institute of Biomedical Science, Fudan University , Shanghai , China.,c Key Laboratory of Molecular Medicine, Ministry of Education Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Duan Ma
- b Institute of Biomedical Science, Fudan University , Shanghai , China.,c Key Laboratory of Molecular Medicine, Ministry of Education Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Xiaotian Li
- a Obstetrics & Gynecology Hospital , Shanghai , China.,b Institute of Biomedical Science, Fudan University , Shanghai , China.,d Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , China
| |
Collapse
|
41
|
Rätsep MT, Paolozza A, Hickman AF, Maser B, Kay VR, Mohammad S, Pudwell J, Smith GN, Brien D, Stroman PW, Adams MA, Reynolds JN, Croy BA, Forkert ND. Brain Structural and Vascular Anatomy Is Altered in Offspring of Pre-Eclamptic Pregnancies: A Pilot Study. AJNR Am J Neuroradiol 2015; 37:939-45. [PMID: 26721772 DOI: 10.3174/ajnr.a4640] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/05/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE Pre-eclampsia is a serious clinical gestational disorder occurring in 3%-5% of all human pregnancies and characterized by endothelial dysfunction and vascular complications. Offspring born of pre-eclamptic pregnancies are reported to exhibit deficits in cognitive function, higher incidence of depression, and increased susceptibility to stroke. However, no brain imaging reports exist on these offspring. We aimed to assess brain structural and vascular anatomy in 7- to 10-year-old offspring of pre-eclamptic pregnancies compared with matched controls. MATERIALS AND METHODS Offspring of pre-eclamptic pregnancies and matched controls (n = 10 per group) were recruited from an established longitudinal cohort examining the effects of pre-eclampsia. Children underwent MR imaging to identify brain structural and vascular anatomic differences. Maternal plasma samples collected at birth were assayed for angiogenic factors by enzyme-linked immunosorbent assay. RESULTS Offspring of pre-eclamptic pregnancies exhibited enlarged brain regional volumes of the cerebellum, temporal lobe, brain stem, and right and left amygdalae. These offspring displayed reduced cerebral vessel radii in the occipital and parietal lobes. Enzyme-linked immunosorbent assay analysis revealed underexpression of the placental growth factor among the maternal plasma samples from women who experienced pre-eclampsia. CONCLUSIONS This study is the first to report brain structural and vascular anatomic alterations in the population of offspring of pre-eclamptic pregnancies. Brain structural alterations shared similarities with those seen in autism. Vascular alterations may have preceded these structural alterations. This pilot study requires further validation with a larger population to provide stronger estimates of brain structural and vascular outcomes among the offspring of pre-eclamptic pregnancies.
Collapse
Affiliation(s)
- M T Rätsep
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.)
| | - A Paolozza
- Centre for Neuroscience Studies (A.P., D.B., P.W.S., M.A.A., J.N.R.), Queen's University, Kingston, Ontario, Canada
| | - A F Hickman
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.)
| | - B Maser
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.)
| | - V R Kay
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.)
| | - S Mohammad
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.)
| | - J Pudwell
- Department of Obstetrics and Gynecology (J.P., G.N.S.), Kingston General Hospital, Kingston, Ontario, Canada
| | - G N Smith
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.) Department of Obstetrics and Gynecology (J.P., G.N.S.), Kingston General Hospital, Kingston, Ontario, Canada
| | - D Brien
- Centre for Neuroscience Studies (A.P., D.B., P.W.S., M.A.A., J.N.R.), Queen's University, Kingston, Ontario, Canada
| | - P W Stroman
- Centre for Neuroscience Studies (A.P., D.B., P.W.S., M.A.A., J.N.R.), Queen's University, Kingston, Ontario, Canada
| | - M A Adams
- Centre for Neuroscience Studies (A.P., D.B., P.W.S., M.A.A., J.N.R.), Queen's University, Kingston, Ontario, Canada
| | - J N Reynolds
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.) Centre for Neuroscience Studies (A.P., D.B., P.W.S., M.A.A., J.N.R.), Queen's University, Kingston, Ontario, Canada
| | - B A Croy
- From the Department of Biomedical and Molecular Sciences (M.T.R., A.F.H., B.M., V.R.K., S.M., G.N.S., J.N.R., B.A.C.)
| | - N D Forkert
- Department of Radiology and Hotchkiss Brain Institute (N.D.F.), University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|