1
|
Wu P, Shen N, Feng S, Liu W, Wang J, Wang C. Oxidative stress and apoptosis of the spinal cord in a rat model of retinoic acid-induced neural tube defects. Int J Dev Neurosci 2025; 85:e10399. [PMID: 39633511 DOI: 10.1002/jdn.10399] [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: 08/31/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Neural tube defects (NTDs) are severe congenital anomalies that significantly impact the central nervous system, arising from the neural tube's failure to close during early embryogenesis. In this study, we investigated NTDs and associated pathophysiological mechanisms in foetal rats following exposure to all-trans retinoic acid (atRA). Out of 168 embryos from 15 pregnant rats in the experimental group, 78% displayed NTDs with notable spinal deformities, primarily in the lumbar-sacral region, similar to human cases. Body weight and crown-rump length (CRL) measurements indicated significant growth impairment in the NTD group compared to controls, while the atRA-treated group without NTDs showed no notable differences in growth. Immunohistochemistry (IHC) results demonstrated decreased NeuN and PCNA expression in the NTD group's spinal cord. Oxidative stress markers showed markedly reduced superoxide dismutase (SOD) and glutathione peroxidase (GSH-px) activity, alongside increased malondialdehyde (MDA) levels in the NTD group, indicating heightened oxidative stress. Analysis of apoptosis-related proteins revealed elevated Bax and caspase-3 levels, reduced Bcl-2 and lower poly (ADP-ribose) polymerase (PARP) in the NTD group, suggesting a pronounced shift towards proapoptotic pathways, potentially contributing to NTD progression. Our findings indicate that oxidative stress and apoptosis play significant roles in the development of NTDs. Future investigations should aim to pinpoint critical regulatory genes or proteins that might be targeted for therapeutic interventions to alleviate oxidative stress and apoptosis in NTD development.
Collapse
Affiliation(s)
- Peng Wu
- Department of Pediatric Surgery, Northwest Women and Children's Hospital, Xi'an, China
| | - Nan Shen
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, China
| | - Shaoguang Feng
- Hangzhou Children's Hospital, Department of Pediatric Surgery, Hangzhou, China
| | - Weiguang Liu
- Hangzhou Children's Hospital, Department of Pediatric Surgery, Hangzhou, China
| | - Jun Wang
- Hangzhou Children's Hospital, Department of Pediatric Surgery, Hangzhou, China
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Wang
- Department of Pediatric Surgery, Hangzhou Children's Hospital, Zhejiang, Hangzhou, China
| |
Collapse
|
2
|
Aguayo-Gómez A, Luna-Muñoz L, Svyryd Y, Muñoz-Téllez LÁ, Mutchinick OM. Bayesian polygenic risk estimation approach to nuclear families with discordant sib-pairs for myelomeningocele. PLoS One 2024; 19:e0316378. [PMID: 39774454 PMCID: PMC11684611 DOI: 10.1371/journal.pone.0316378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Myelomeningocele (MMC) is the most severe and disabling form of spina bifida with chronic health multisystem complications and social and economic family and health systems burden. In the present study, we aimed to investigate the genetic risk estimate for MMC in a cohort of 203 Mexican nuclear families with discordant siblings for the defect. Utilizing a custom Illumina array, we analyzed 656 single nucleotide polymorphisms (SNPs) of 395 candidate genes to identify a polygenic risk profile for MMC. Through a family-based analysis employing the transmission disequilibrium test (TDT) and Bayesian analysis, we assessed risk alleles transmission and calculated conditional probabilities estimating a polygenic risk for MMC. Our findings reveal significant associations of six genes related to neural tube closure (PSMB4, ATIC, DKK2, PSEN2, C2CD3, and PLCB2), showing differences in risk allele transmission between affected and unaffected siblings. Bayesian analysis identified changes in the risk profile after initiating folic acid fortification in Mexico, showing an evident decline in the conditional risk from 1/156 to 1/304 respectively. Despite the decline, this represents a 5.84-fold increase in risk before fortification and a 2.99-fold increase post-fortification compared to the baseline risk level (1/910). Our study highlights the advantage of incorporating a Bayesian analytical methodology in families with discordant sib-pairs, offering insights into the polygenic risk estimate for MMC and, most probably, for other congenital malformations.
Collapse
Affiliation(s)
- Adolfo Aguayo-Gómez
- Department of Genetics, National Institute of Medical Sciences and Nutrition Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - Leonora Luna-Muñoz
- Department of Genetics, National Institute of Medical Sciences and Nutrition Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - Yevgeniya Svyryd
- Department of Genetics, National Institute of Medical Sciences and Nutrition Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - Luis Ángel Muñoz-Téllez
- Department of Genetics, National Institute of Medical Sciences and Nutrition Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - Osvaldo M. Mutchinick
- Department of Genetics, National Institute of Medical Sciences and Nutrition Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| |
Collapse
|
3
|
Wang G, Shen WB, Chen AW, Reece EA, Yang P. Diabetes and Early Development: Epigenetics, Biological Stress, and Aging. Am J Perinatol 2024. [PMID: 39209306 DOI: 10.1055/a-2405-1493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pregestational diabetes, either type 1 or type 2 diabetes, induces structural birth defects including neural tube defects and congenital heart defects in human fetuses. Rodent models of type 1 and type 2 diabetic embryopathy have been established and faithfully mimic human conditions. Hyperglycemia of maternal diabetes triggers oxidative stress in the developing neuroepithelium and the embryonic heart leading to the activation of proapoptotic kinases and excessive cell death. Oxidative stress also activates the unfolded protein response and endoplasmic reticulum stress. Hyperglycemia alters epigenetic landscapes by suppressing histone deacetylation, perturbing microRNA (miRNA) expression, and increasing DNA methylation. At cellular levels, besides the induction of cell apoptosis, hyperglycemia suppresses cell proliferation and induces premature senescence. Stress signaling elicited by maternal diabetes disrupts cellular organelle homeostasis leading to mitochondrial dysfunction, mitochondrial dynamic alteration, and autophagy impairment. Blocking oxidative stress, kinase activation, and cellular senescence ameliorates diabetic embryopathy. Deleting the mir200c gene or restoring mir322 expression abolishes maternal diabetes hyperglycemia-induced senescence and cellular stress, respectively. Both the autophagy activator trehalose and the senomorphic rapamycin can alleviate diabetic embryopathy. Thus, targeting cellular stress, miRNAs, senescence, or restoring autophagy or mitochondrial fusion is a promising approach to prevent poorly controlled maternal diabetes-induced structural birth defects. In this review, we summarize the causal events in diabetic embryopathy and propose preventions for this pathological condition. KEY POINTS: · Maternal diabetes induces structural birth defects.. · Kinase signaling and cellular organelle stress are critically involved in neural tube defects.. · Maternal diabetes increases DNA methylation and suppresses developmental gene expression.. · Cellular apoptosis and senescence are induced by maternal diabetes in the neuroepithelium.. · microRNAs disrupt mitochondrial fusion leading to congenital heart diseases in diabetic pregnancy..
Collapse
Affiliation(s)
- Guanglei Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anna Wu Chen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - E Albert Reece
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Yan Y, Huang W, Lu X, Chen X, Shan Y, Luo X, Li Y, Yang X, Li C. Zinc oxide nanoparticles induces cell death and consequently leading to incomplete neural tube closure through oxidative stress during embryogenesis. Cell Biol Toxicol 2024; 40:51. [PMID: 38958792 PMCID: PMC11222284 DOI: 10.1007/s10565-024-09894-1] [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: 01/29/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
The implementation of Zinc oxide nanoparticles (ZnO NPs) raises concerns regarding their potential toxic effects on human health. Although more and more researches have confirmed the toxic effects of ZnO NPs, limited attention has been given to their impact on the early embryonic nervous system. This study aimed to explore the impact of exposure to ZnO NPs on early neurogenesis and explore its underlying mechanisms. We conducted experiments here to confirm the hypothesis that exposure to ZnO NPs causes neural tube defects in early embryonic development. We first used mouse and chicken embryos to confirm that ZnO NPs and the Zn2+ they release are able to penetrate the placental barrier, influence fetal growth and result in incomplete neural tube closure. Using SH-SY5Y cells, we determined that ZnO NPs-induced incomplete neural tube closure was caused by activation of various cell death modes, including ferroptosis, apoptosis and autophagy. Moreover, dissolved Zn2+ played a role in triggering widespread cell death. ZnO NPs were accumulated within mitochondria after entering cells, damaging mitochondrial function and resulting in the over production of reactive oxygen species, ultimately inducing cellular oxidative stress. The N-acetylcysteine (NAC) exhibits significant efficacy in mitigating cellular oxidative stress, thereby alleviating the cytotoxicity and neurotoxicity brought about by ZnO NPs. These findings indicated that the exposure of ZnO NPs in early embryonic development can induce cell death through oxidative stress, resulting in a reduced number of cells involved in early neural tube closure and ultimately resulting in incomplete neural tube closure during embryo development. The findings of this study could raise public awareness regarding the potential risks associated with the exposure and use of ZnO NPs in early pregnancy.
Collapse
Affiliation(s)
- Yu Yan
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wenyi Huang
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Research Center of Integrative Medicine, School Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoting Lu
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Research Center of Integrative Medicine, School Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianxian Chen
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Research Center of Integrative Medicine, School Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingyi Shan
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center of Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Xin Luo
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Yu Li
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Xuesong Yang
- Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China.
- Clinical Research Center, Clifford Hospital, Guangzhou, 511495, China.
| | - Chun Li
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
5
|
Yang J, Liu D, Du Q, Zhu J, Lu L, Wu Z, Zhang D, Ji X, Zheng X. Construction of a 3-year risk prediction model for developing diabetes in patients with pre-diabetes. Front Endocrinol (Lausanne) 2024; 15:1410502. [PMID: 38938520 PMCID: PMC11208327 DOI: 10.3389/fendo.2024.1410502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024] Open
Abstract
Introduction To analyze the influencing factors for progression from newly diagnosed prediabetes (PreDM) to diabetes within 3 years and establish a prediction model to assess the 3-year risk of developing diabetes in patients with PreDM. Methods Subjects who were diagnosed with new-onset PreDM at the Physical Examination Center of the First Affiliated Hospital of Soochow University from October 1, 2015 to May 31, 2023 and completed the 3-year follow-up were selected as the study population. Data on gender, age, body mass index (BMI), waist circumference, etc. were collected. After 3 years of follow-up, subjects were divided into a diabetes group and a non-diabetes group. Baseline data between the two groups were compared. A prediction model based on logistic regression was established with nomogram drawn. The calibration was also depicted. Results Comparison between diabetes group and non-diabetes group: Differences in 24 indicators including gender, age, history of hypertension, fatty liver, BMI, waist circumference, systolic blood pressure, diastolic blood pressure, fasting blood glucose, HbA1c, etc. were statistically significant between the two groups (P<0.05). Differences in smoking, creatinine and platelet count were not statistically significant between the two groups (P>0.05). Logistic regression analysis showed that ageing, elevated BMI, male gender, high fasting blood glucose, increased LDL-C, fatty liver, liver dysfunction were risk factors for progression from PreDM to diabetes within 3 years (P<0.05), while HDL-C was a protective factor (P<0.05). The derived formula was: In(p/1-p)=0.181×age (40-54 years old)/0.973×age (55-74 years old)/1.868×age (≥75 years old)-0.192×gender (male)+0.151×blood glucose-0.538×BMI (24-28)-0.538×BMI (≥28)-0.109×HDL-C+0.021×LDL-C+0.365×fatty liver (yes)+0.444×liver dysfunction (yes)-10.038. The AUC of the model for predicting progression from PreDM to diabetes within 3 years was 0.787, indicating good predictive ability of the model. Conclusions The risk prediction model for developing diabetes within 3 years in patients with PreDM constructed based on 8 influencing factors including age, BMI, gender, fasting blood glucose, LDL-C, HDL-C, fatty liver and liver dysfunction showed good discrimination and calibration.
Collapse
Affiliation(s)
- Jianshu Yang
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dan Liu
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiaoqiao Du
- Health Management Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jing Zhu
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Lu
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengyan Wu
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Daiyi Zhang
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaodong Ji
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Zheng
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Wang G, Lu W, Shen WB, Karbowski M, Kaushal S, Yang P. Small Molecule Activators of Mitochondrial Fusion Prevent Congenital Heart Defects Induced by Maternal Diabetes. JACC Basic Transl Sci 2024; 9:303-318. [PMID: 38559623 PMCID: PMC10978414 DOI: 10.1016/j.jacbts.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 04/04/2024]
Abstract
Most congenital heart defect (CHD) cases are attributed to nongenetic factors; however, the mechanisms underlying nongenetic factor-induced CHDs are elusive. Maternal diabetes is one of the nongenetic factors, and this study aimed to determine whether impaired mitochondrial fusion contributes to maternal diabetes-induced CHDs and if mitochondrial fusion activators, teriflunomide and echinacoside, could reduce CHD incidence in diabetic pregnancy. We demonstrated maternal diabetes-activated FoxO3a increases miR-140 and miR-195, which in turn represses Mfn1 and Mfn2, leading to mitochondrial fusion defects and CHDs. Two mitochondrial fusion activators are effective in preventing CHDs in diabetic pregnancy.
Collapse
Affiliation(s)
- Guanglei Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wenhui Lu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mariusz Karbowski
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sunjay Kaushal
- Division of Cardiac Surgery, Department of Surgery, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Nishino T, Ranade SS, Pelonero A, van Soldt BJ, Ye L, Alexanian M, Koback F, Huang Y, Sadagopan N, Lam A, Zholudeva LV, Li F, Padmanabhan A, Thomas R, van Bemmel JG, Gifford CA, Costa MW, Srivastava D. Single Cell Multimodal Analyses Reveal Epigenomic and Transcriptomic Basis for Birth Defects in Maternal Diabetes. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1190-1203. [PMID: 39183978 PMCID: PMC11343316 DOI: 10.1038/s44161-023-00367-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/19/2023] [Indexed: 08/27/2024]
Abstract
Maternal diabetes mellitus is among the most frequent environmental contributors to congenital birth defects, including heart defects and craniofacial anomalies, yet the cell types affected and mechanisms of disruption are largely unknown. Using multi-modal single cell analyses, here we show that maternal diabetes affects the epigenomic landscape of specific subsets of cardiac and craniofacial progenitors during embryogenesis. A previously unrecognized cardiac progenitor subpopulation expressing the homeodomain-containing protein ALX3 showed prominent chromatin accessibility changes and acquired a more posterior identity. Similarly, a subpopulation of neural crest-derived cells in the second pharyngeal arch, which contributes to craniofacial structures, displayed abnormalities in the epigenetic landscape and axial patterning defects. Chromatin accessibility changes in both populations were associated with increased retinoic acid signaling, known to establish anterior-posterior identity. This work highlights how an environmental insult can have highly selective epigenomic consequences on discrete cell types leading to developmental patterning defects.
Collapse
Affiliation(s)
- Tomohiro Nishino
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Sanjeev S. Ranade
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Angelo Pelonero
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Benjamin J. van Soldt
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Lin Ye
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Michael Alexanian
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Frances Koback
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Yu Huang
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Nandhini Sadagopan
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
- Division of Cardiology, Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Adrienne Lam
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Lyandysha V. Zholudeva
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Feiya Li
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Arun Padmanabhan
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
- Division of Cardiology, Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | | | - Joke G. van Bemmel
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Casey A. Gifford
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Mauro W. Costa
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Deepak Srivastava
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
- Division of Cardiology, Department of Pediatrics, University of California, San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco; San Francisco, CA, USA
| |
Collapse
|
8
|
Wang X, Yang C, Ru Y, Xie L, Xiao B, Jin X, Ma C, Chai Z, Fan H. An optimal combination of five main monomer components in Wuzi Yanzong Pill that prevents neural tube defects and reduces apoptosis and oxidative stress. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116540. [PMID: 37088238 DOI: 10.1016/j.jep.2023.116540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wuzi Yanzong pill (WYP) is a classic traditional Chinese medicine (TCM) formula that is used for reproductive system diseases. Previous studies showed that WYP had a preventive effect on the development of neural tube defects (NTDs) induced by all-trans retinoic acid (atRA) in mice. AIM OF THE STUDY This study aimed to determine the optimal combination of main monomer components in WYP on preventing NTDs and to understand the underlying mechanism. MATERIALS AND METHODS An optimal combination was made from five representative components in WYP including hyperoside, acteoside, schizandrol A, kaempferide and ellagic acid by orthogonal design method. In a mouse model of NTDs induced by intraperitoneal injection of atRA, pathological changes of neural tube tissues were observed by Hematoxylin & Eosin (HE) staining, neural tube epithelial cells apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL), protein changes related to apoptosis, anti-apoptosis, and antioxidant factors were detected with Western blot. Potential targets and mechanisms of monomer compatibility group (MCG) acting on NTDs were analyzed by bioinformatics. RESULTS Treatment with different combinations of WYP bioactive ingredients resulted in varying decreases in the incidence of NTDs in mice embryos. The combination of MCG15 (200 mg/kg of hyperoside, 100 mg/kg of acteoside, 10 mg/kg of schizandrol A, 100 mg/kg of kaempferide and 1 mg/kg of ellagic acid) showed the most significant reduction in NTD incidence. Mechanistically, MCG15 inhibited apoptosis and oxidative stress, as evidenced by reduced TUNEL-positive cells, downregulation of caspase-9, cleaved caspase-3, Bad, and Bax, and upregulation of Bcl-2, as well as decreased MDA and increased SOD, CAT, GSH, HO-1, and GPX1 levels. Bioinformatics analysis showed that MCG15 acted on the PI3K/Akt signaling pathway, which was confirmed by Western blot analysis showing increased expression of p-PI3K, p-Akt/Akt, and Nrf2 related indicators. CONCLUSION We have identified an optimal combination of five bioactive components in WYP (MCG15) that prevented NTDs in mice embryos induced by atRA by activating the PI3K/Akt signaling pathway and inhibiting apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Xinliang Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Neurobiology Research Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Chanjuan Yang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Neurobiology Research Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yi Ru
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Neurobiology Research Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Liangqi Xie
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Neurobiology Research Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Baoguo Xiao
- Huashan Hospital, Fudan University, Shanghai, 200025, China
| | - Xiaoming Jin
- Department of Anatomy and Cell Biology, Department of Neurological Surgery, Stark Neurosciences Research Institute. Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Neurobiology Research Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| | - Zhi Chai
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Neurobiology Research Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| | - Huijie Fan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Neurobiology Research Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| |
Collapse
|
9
|
Klein A, Rhinn M, Keyes WM. Cellular senescence and developmental defects. FEBS J 2023; 290:1303-1313. [PMID: 36856681 DOI: 10.1111/febs.16731] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/05/2023] [Accepted: 01/20/2023] [Indexed: 03/02/2023]
Abstract
Cellular senescence is a distinct state that is frequently induced in response to ageing and stress. Yet studies have also uncovered beneficial functions in development, repair and regeneration. Current opinion therefore suggests that timely and controlled induction of senescence can be beneficial, while misregulation of the senescence program, either through mis-timed activation, or chronic accumulation of senescent cells, contributes to many disease states and the ageing process. Whether atypical activation of senescence plays a role in the pathogenesis of developmental defects has been relatively underexplored. Here, we discuss three recent studies that implicate ectopic senescence in neurodevelopmental defects, with possible causative roles for senescence in these birth defects. In addition, we highlight how the examination of senescence in other birth defects is warranted, and speculate that aberrantly activated senescence may play a much broader role in developmental defects than currently appreciated.
Collapse
Affiliation(s)
- Annabelle Klein
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Muriel Rhinn
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - William M Keyes
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France.,Université de Strasbourg, Illkirch, France
| |
Collapse
|
10
|
Engelhardt DM, Martyr CA, Niswander L. Pathogenesis of neural tube defects: The regulation and disruption of cellular processes underlying neural tube closure. WIREs Mech Dis 2022; 14:e1559. [PMID: 35504597 PMCID: PMC9605354 DOI: 10.1002/wsbm.1559] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 11/08/2022]
Abstract
Neural tube closure (NTC) is crucial for proper development of the brain and spinal cord and requires precise morphogenesis from a sheet of cells to an intact three-dimensional structure. NTC is dependent on successful regulation of hundreds of genes, a myriad of signaling pathways, concentration gradients, and is influenced by epigenetic and environmental cues. Failure of NTC is termed a neural tube defect (NTD) and is a leading class of congenital defects in the United States and worldwide. Though NTDs are all defined as incomplete closure of the neural tube, the pathogenesis of an NTD determines the type, severity, positioning, and accompanying phenotypes. In this review, we survey pathogenesis of NTDs relating to disruption of cellular processes arising from genetic mutations, altered epigenetic regulation, and environmental influences by micronutrients and maternal condition. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Engelhardt
- Molecular Cellular Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Cara A Martyr
- Molecular Cellular Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Lee Niswander
- Molecular Cellular Developmental Biology, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
11
|
Cao S, Wu Y, Albert Reece E, Xu C, Shen WB, Kaushal S, Yang P. Functional cargos of exosomes derived from Flk-1 + vascular progenitors enable neurulation and ameliorate embryonic anomalies in diabetic pregnancy. Commun Biol 2022; 5:648. [PMID: 35778435 PMCID: PMC9249756 DOI: 10.1038/s42003-022-03614-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 06/21/2022] [Indexed: 11/27/2022] Open
Abstract
Various types of progenitors initiate individual organ formation and their crosstalk orchestrates morphogenesis for the entire embryo. Here we show that progenitor exosomal communication across embryonic organs occurs in normal development and is altered in embryos of diabetic pregnancy. Endoderm fibroblast growth factor 2 (FGF2) stimulates mesoderm Flk-1+ vascular progenitors to produce exosomes containing the anti-stress protein Survivin. These exosomes act on neural stem cells of the neuroepithelium to facilitate neurulation by inhibiting cellular stress and apoptosis. Maternal diabetes causes Flk-1+ progenitor dysfunction by suppressing FGF2 through DNA hypermethylation. Restoring endoderm FGF2 prevents diabetes-induced survivin reduction in Flk-1+ progenitor exosomes. Transgenic Survivin expression in Flk-1+ progenitors or in utero delivery of survivin-enriched exosomes restores cellular homeostasis and prevents diabetes-induced neural tube defects (NTDs), whereas inhibiting exosome production induces NTDs. Thus, functional inter-organ communication via Flk-1 exosomes is vital for neurulation and its disruption leads to embryonic anomalies.
Collapse
Affiliation(s)
- Songying Cao
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yanqing Wu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute of Life Sciences, Wenzhou University, Zhejiang Province, 325035, Wenzhou, China
| | - E Albert Reece
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cheng Xu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sunjay Kaushal
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Cardiovascular-Thoracic Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E. Chicago Avenue, Chicago, IL, 60611, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Nuclear factor I-C disrupts cellular homeostasis between autophagy and apoptosis via miR-200b-Ambra1 in neural tube defects. Cell Death Dis 2021; 13:17. [PMID: 34930914 PMCID: PMC8688449 DOI: 10.1038/s41419-021-04473-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/25/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023]
Abstract
Impaired autophagy and excessive apoptosis disrupt cellular homeostasis and contribute to neural tube defects (NTDs), which are a group of fatal and disabling birth defects caused by the failure of neural tube closure during early embryonic development. However, the regulatory mechanisms underlying NTDs and outcomes remain elusive. Here, we report the role of the transcription factor nuclear factor I-C (NFIC) in maintaining cellular homeostasis in NTDs. We demonstrated that abnormally elevated levels of NFIC in a mouse model of NTDs can interact with the miR-200b promoter, leading to the activation of the transcription of miR-200b, which plays a critical role in NTD formation, as reported in our previous study. Furthermore, miR-200b represses autophagy and triggers apoptosis by directly targeting the autophagy-related gene Ambra1 (Autophagy/Beclin1 regulator 1). Notably, miR-200b inhibitors mitigate the unexpected effects of NFIC on autophagy and apoptosis. Collectively, these results indicate that the NFIC-miR-200b-Ambra1 axis, which integrates transcription- and epigenome-regulated miRNAs and an autophagy regulator, disrupts cellular homeostasis during the closure of the neural tube, and may provide new insight into NTD pathogenesis.
Collapse
|
13
|
Choudhury TZ, Majumdar U, Basu M, Garg V. Impact of maternal hyperglycemia on cardiac development: Insights from animal models. Genesis 2021; 59:e23449. [PMID: 34498806 PMCID: PMC8599640 DOI: 10.1002/dvg.23449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022]
Abstract
Congenital heart disease (CHD) is the leading cause of birth defect-related death in infants and is a global pediatric health concern. While the genetic causes of CHD have become increasingly recognized with advances in genome sequencing technologies, the etiology for the majority of cases of CHD is unknown. The maternal environment during embryogenesis has a profound impact on cardiac development, and numerous environmental factors are associated with an elevated risk of CHD. Maternal diabetes mellitus (matDM) is associated with up to a fivefold increased risk of having an infant with CHD. The rising prevalence of diabetes mellitus has led to a growing interest in the use of experimental diabetic models to elucidate mechanisms underlying this associated risk for CHD. The purpose of this review is to provide a comprehensive summary of rodent models that are being used to investigate alterations in cardiac developmental pathways when exposed to a maternal diabetic setting and to summarize the key findings from these models. The majority of studies in the field have utilized the chemically induced model of matDM, but recent advances have also been made using diet based and genetic models. Each model provides an opportunity to investigate unique aspects of matDM and is invaluable for a comprehensive understanding of the molecular and cellular mechanisms underlying matDM-associated CHD.
Collapse
Affiliation(s)
- Talita Z. Choudhury
- Center for Cardiovascular Research and Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, United States
- Graduate Program in Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH 43210, United States
| | - Uddalak Majumdar
- Center for Cardiovascular Research and Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, United States
| | - Madhumita Basu
- Center for Cardiovascular Research and Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, United States
| | - Vidu Garg
- Center for Cardiovascular Research and Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, United States
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
14
|
Nakano H, Fajardo VM, Nakano A. The role of glucose in physiological and pathological heart formation. Dev Biol 2021; 475:222-233. [PMID: 33577830 PMCID: PMC8107118 DOI: 10.1016/j.ydbio.2021.01.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/30/2020] [Accepted: 01/29/2021] [Indexed: 02/08/2023]
Abstract
Cells display distinct metabolic characteristics depending on its differentiation stage. The fuel type of the cells serves not only as a source of energy but also as a driver of differentiation. Glucose, the primary nutrient to the cells, is a critical regulator of rapidly growing embryos. This metabolic change is a consequence as well as a cause of changes in genetic program. Disturbance of fetal glucose metabolism such as in diabetic pregnancy is associated with congenital heart disease. In utero hyperglycemia impacts the left-right axis establishment, migration of cardiac neural crest cells, conotruncal formation and mesenchymal formation of the cardiac cushion during early embryogenesis and causes cardiac hypertrophy in late fetal stages. In this review, we focus on the role of glucose in cardiogenesis and the molecular mechanisms underlying heart diseases associated with hyperglycemia.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Viviana M Fajardo
- Department of Pediatrics, Division of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
15
|
Xu C, Shen WB, Reece EA, Hasuwa H, Harman C, Kaushal S, Yang P. Maternal diabetes induces senescence and neural tube defects sensitive to the senomorphic rapamycin. SCIENCE ADVANCES 2021; 7:7/27/eabf5089. [PMID: 34193422 PMCID: PMC8245044 DOI: 10.1126/sciadv.abf5089] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/18/2021] [Indexed: 05/03/2023]
Abstract
Neural tube defects (NTDs) are the second most common structural birth defect. Senescence, a state of permanent cell cycle arrest, occurs only after neural tube closure. Maternal diabetes-induced NTDs are severe diabetic complications that lead to infant mortality or lifelong morbidity and may be linked to premature senescence. Here, we report that premature senescence occurs in the mouse neuroepithelium and disrupts neurulation, leading to NTDs in diabetic pregnancy. Premature senescence and NTDs were abolished by knockout of the transcription factor Foxo3a, the miR-200c gene, and the cell cycle inhibitors p21 and p27; transgenic expression of the dominant-negative FoxO3a mutant; or the senomorphic rapamycin. Double transgenic expression of p21 and p27 mimicked maternal diabetes in inducing premature neuroepithelium senescence and NTDs. These findings integrate transcription- and epigenome-regulated miRNAs and cell cycle regulators in premature neuroepithelium senescence and provide a mechanistic basis for targeting premature senescence and NTDs using senomorphics.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - E Albert Reece
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hidetoshi Hasuwa
- Department of Molecular Biology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| | - Christopher Harman
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sunjay Kaushal
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Mor ME, Harvey A, Familari M, St Clair-Glover M, Viventi S, de Iongh RU, Cameron FJ, Dottori M. Neural differentiation medium for human pluripotent stem cells to model physiological glucose levels in human brain. Brain Res Bull 2021; 173:141-149. [PMID: 34022288 DOI: 10.1016/j.brainresbull.2021.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 05/05/2021] [Accepted: 05/17/2021] [Indexed: 12/30/2022]
Abstract
Cortical neurospheres (NSPs) derived from human pluripotent stem cells (hPSC), have proven to be a successful platform to investigate human brain development and neuro-related diseases. Currently, many of the standard hPSC neural differentiation media, use concentrations of glucose (approximately 17.5-25 mM) and insulin (approximately 3.2 μM) that are much greater than the physiological concentrations found in the human brain. These culture conditions make it difficult to analyse perturbations of glucose or insulin on neuronal development and differentiation. We established a new hPSC neural differentiation medium that incorporated physiological brain concentrations of glucose (2.5 mM) and significantly reduced insulin levels (0.86 μM). This medium supported hPSC neural induction and formation of cortical NSPs. The revised hPSC neural differentiation medium, may provide an improved platform to model brain development and to investigate neural differentiation signalling pathways impacted by abnormal glucose and insulin levels.
Collapse
Affiliation(s)
- Michal E Mor
- Department of Anatomy & Physiology, University of Melbourne, Australia
| | | | - Mary Familari
- School of BioSciences, University of Melbourne, Australia
| | - Mitchell St Clair-Glover
- Illawarra Health and Medical Research Institute, Molecular Horizons, University of Wollongong, Australia
| | - Serena Viventi
- The Florey Institute of Neuroscience and Mental Health, Australia
| | - Robb U de Iongh
- Department of Anatomy & Physiology, University of Melbourne, Australia
| | - Fergus J Cameron
- Murdoch Children's Research Institute, The Royal Children's Hospital, Australia; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Australia
| | - Mirella Dottori
- Department of Anatomy & Physiology, University of Melbourne, Australia; Illawarra Health and Medical Research Institute, Molecular Horizons, University of Wollongong, Australia; Department of Biomedical Engineering, University of Melbourne, Australia.
| |
Collapse
|
17
|
Li T, Hu D, Gong Y. Identification of potential lncRNAs and co-expressed mRNAs in gestational diabetes mellitus by RNA sequencing. J Matern Fetal Neonatal Med 2021; 35:5125-5139. [PMID: 33618585 DOI: 10.1080/14767058.2021.1875432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
AIM Gestational diabetes mellitus is common during pregnancy, impacting maternal health and fetal development. The aim of this study was to identify potential long non-coding RNAs (lncRNAs) and mRNAs in gestational diabetes mellitus. METHODS The placenta tissues from four women patients with gestational diabetes mellitus and three healthy pregnant women were used for RNA sequencing. Differentially expressed lncRNAs and mRNAs were obtained. Then, interaction networks of lncRNA-nearby targeted mRNA and lncRNA-co-expressed mRNA were constructed, followed by functional annotation of co-expressed mRNAs. Third, GSE51546 dataset was utilized to validate the expression of selected co-expressed mRNAs. In addition, in vitro experiment was applied to expression validation of lncRNAs and mRNAs. Finally, GSE70493 dataset was utilized for diagnostic analysis of selected co-expressed mRNAs. RESULTS A total of 78 differentially expressed lncRNAs and 647 differentially expressed mRNAs in gestational diabetes mellitus were obtained. Several interaction pairs of lncRNA-co-expressed mRNA including LINC01504-CASP8, FUT8-AS1-TLR5/GDF15, GATA2-AS1-PQLC3/KIAA2026, and EGFR-AS1-HLA-G were identified. Endocytosis (involved HLA-G) and toll-like receptor signaling pathway (involved TLR5 and CASP8) were remarkably enriched signaling pathways of co-expressed mRNAs. It is noted that CASP8, TLR5, and PQLC3 had a significant prognosis value for gestational diabetes mellitus. CONCLUSIONS Our study identified several differentially expressed lncRNAs and mRNAs, and their interactions, especially co-expression, may be associated with gestational diabetes mellitus.
Collapse
Affiliation(s)
- Tao Li
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, P. R. China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, P. R. China
| | - Die Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, P. R. China.,Department of Outpatient, West China Second University Hospital, Sichuan University, Chengdu, P. R. China
| | - Yunhui Gong
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, P. R. China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
18
|
Fitzgerald E, Hor K, Drake AJ. Maternal influences on fetal brain development: The role of nutrition, infection and stress, and the potential for intergenerational consequences. Early Hum Dev 2020; 150:105190. [PMID: 32948364 PMCID: PMC7481314 DOI: 10.1016/j.earlhumdev.2020.105190] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
An optimal early life environment is crucial for ensuring ideal neurodevelopmental outcomes. Brain development consists of a finely tuned series of spatially and temporally constrained events, which may be affected by exposure to a sub-optimal intra-uterine environment. Evidence suggests brain development may be particularly vulnerable to factors such as maternal nutrition, infection and stress during pregnancy. In this review, we discuss how maternal factors such as these can affect brain development and outcome in offspring, and we also identify evidence which suggests that the outcome can, in many cases, be stratified by socio-economic status (SES), with individuals in lower brackets typically having a worse outcome. We consider the relevant epidemiological evidence and draw parallels to mechanisms suggested by preclinical work where appropriate. We also discuss possible transgenerational effects of these maternal factors and the potential mechanisms involved. We conclude that modifiable factors such as maternal nutrition, infection and stress are important contributors to atypical brain development and that SES also likely has a key role.
Collapse
Affiliation(s)
- Eamon Fitzgerald
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Kahyee Hor
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Amanda J Drake
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
19
|
Cao S, Shen WB, Reece EA, Yang P. Deficiency of the oxidative stress-responsive kinase p70S6K1 restores autophagy and ameliorates neural tube defects in diabetic embryopathy. Am J Obstet Gynecol 2020; 223:753.e1-753.e14. [PMID: 32416155 PMCID: PMC7609618 DOI: 10.1016/j.ajog.2020.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Autophagy is highly active in neuroepithelial cells of the developing neuroepithelium, and impairment of autophagy leads to neural tube defects. In this study, we have found that maternal diabetes suppresses autophagy that leads to neural tube defects and consequent cellular imbalance in the endoplasmic reticulum where critical events occur, leading to the induction of diabetic embryopathy. Because the mammalian target of rapamycin pathway suppresses autophagy, we hypothesized that 70 kDa ribosomal protein S6 kinase 1 (p70S6K1), a major downstream effector of mammalian target of rapamycin, mediates the inhibitory effect of maternal diabetes on autophagy in the developing neuroepithelium. OBJECTIVE We investigated whether p70S6K1 mediates the inhibitory effect of maternal diabetes on autophagy during neurulation. We also examined whether p70S6K1 deficiency restores autophagy and therefore relieves endoplasmic reticulum stress and inhibits maternal diabetes-induced apoptosis, which leads to reduction in neural tube defect incidence in diabetic embryopathy. STUDY DESIGN Female p70S6K1 heterogeneous knockout (p70S6K1+/-) mice were bred with male p70S6K1 heterogeneous knockout (p70S6K1+/-) mice to generate wild-type (WT), p70S6K1+/- and p70S6K1 knockout (p70S6K1-/-) embryos. Embryos at embryonic day 8.5 were harvested for the assessment of indices of autophagy, endoplasmic reticulum stress, and apoptosis. Neural tube defect incidence in embryos was determined at embryonic day 10.5. For in vitro studies, small interfering RNA knockdown of p70S6K1 in C17.2 mouse neural stem cells was used to determine the effect of p70S6K1 deficiency on autophagy impairment and endoplasmic reticulum stress under high glucose conditions. RESULTS Knockout of the Rps6kb1 gene, which encodes for p70S6K1, ameliorated maternal diabetes-induced NTDs and restored autophagosome formation in neuroepithelial cells suppressed by maternal diabetes. Maternal diabetes-suppressed conversion of LC3-I (microtubule-associated protein 1A/1B-light chain 3) to LC3-II, an index of autophagic activity, in neurulation stage embryos was abrogated in the absence of p70S6K1. p70S6K1 knockdown in neural stem cells also restored autophagosome formation and the conversion of LC3-I to LC3-II. The activation of the major unfolded protein response, indicated by phosphorylation of inositol-requiring enzyme 1 alpha, and protein kinase R-like endoplasmic reticulum kinase, and eukaryotic translation initiation factor 2α, and the increase of the endoplasmic reticulum stress marker, C/EBP homologous protein, were induced by maternal diabetes in vivo and high glucose in vitro. Unfolded protein response and endoplasmic reticulum stress induced by maternal diabetes or high glucose were reduced by Rps6kb1 deletion or p70S6K1 knockdown, respectively. Rps6kb1 knockout blocked maternal diabetes-induced caspase cleavage and neuroepithelial cell apoptosis. The superoxide dismutase mimetic Tempol abolished high glucose-induced p70S6K1 activation. CONCLUSION The study revealed the critical involvement of p70S6K1 in the pathogenesis of diabetic embryopathy.
Collapse
Affiliation(s)
- Songying Cao
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Wei-Bin Shen
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - E Albert Reece
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Peixin Yang
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
20
|
Cao S, Reece EA, Shen WB, Yang P. Restoring BMP4 expression in vascular endothelial progenitors ameliorates maternal diabetes-induced apoptosis and neural tube defects. Cell Death Dis 2020; 11:859. [PMID: 33060561 PMCID: PMC7562732 DOI: 10.1038/s41419-020-03078-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/26/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
During mouse embryonic development, vasculogenesis initially occurs in the yolk sac, preceding neurulation. Our previous studies have demonstrated that maternal diabetes induces embryonic vasculopathy at early embryonic developmental stage by suppressing the expression of vascular growth factors including BMP4 (bone morphogenetic protein 4). This study aimed to determine whether restoring diabetes-inhibited BMP4 expression in Flk-1+ progenitors effectively prevented maternal diabetes-induced embryonic vasculopathy and NTDs. Transgenic (Tg) BMP4 expression in the vascular endothelial growth factor receptor 2 (Flk-1)-positive (Flk-1+) progenitors was achieved by crossing a Floxed BMP4 Tg mouse line with the Flk-1-Cre mouse line. Non-BMP4 Tg and BMP4 Tg embryos were harvested at E8.5 to assess the expression of BMP4, markers of endoplasmic reticulum stress, and expression of the Id genes, direct targets of BMP4; and the presence of cleaved caspase 3 and 8, apoptosis, and Smad signaling. BMP4 Tg overexpression neutralized its down-regulation by maternal diabetes in E8.5 embryos. Maternal diabetes-induced Flk-1+ progenitor apoptosis, impairment of blood island formation, and reduction of Flk-1+ progenitor number and blood vessel density, which were reversed by BMP4 Tg expression. BMP4 Tg expression in Flk-1+ progenitors blocked maternal diabetes-induced vasculopathy in early stage embryos (E7.5-E8.5) and consequently led to amelioration of maternal diabetes-induced neural tube defects (NTDs) at E10.5. BMP4 Tg expression inhibited maternal diabetes-induced endoplasmic reticulum stress and caspase cascade activation in the developing neuroepithelium, and reduced neuroepithelial cell apoptosis. BMP4 Tg expression re-activated Smad1/5/8 phosphorylation and reversed maternal diabetes-suppressed Smad4 expression. BMP4 Tg expression restored Id1 and Smad6 expression inhibited by maternal diabetes. In vitro, recombinant BMP4 protein blocked high glucose-induced Flk-1+ progenitor apoptosis and NTDs. These data demonstrate that BMP4 down-regulation in Flk-1+ progenitors are responsible for diabetes-induced yolk sac vasculopathy, and that restoring BMP4 expression prevents vasculopathy and rescues neuroepithelial cells from cellular organelle stress, leading to NTD reduction.
Collapse
Affiliation(s)
- Songying Cao
- Department of Obstetrics, Gynecology & Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - E Albert Reece
- Department of Obstetrics, Gynecology & Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology & Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Science, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
21
|
Liu YS, Gu H, Huang TC, Wei XW, Ma W, Liu D, He YW, Luo WT, Huang JT, Zhao D, Jia SS, Wang F, Zhang T, Bai YZ, Wang WL, Yuan ZW. miR-322 treatment rescues cell apoptosis and neural tube defect formation through silencing NADPH oxidase 4. CNS Neurosci Ther 2020; 26:902-912. [PMID: 32329577 PMCID: PMC7415201 DOI: 10.1111/cns.13383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/31/2020] [Accepted: 04/05/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS Failure of neural tube closure resulting from excessive apoptosis leads to neural tube defects (NTDs). NADPH oxidase 4 (NOX4) is a critical mediator of cell growth and death, yet its role in NTDs has never been characterized. NOX4 is a potential target of miR-322, and we have previously demonstrated that miR-322 was involved in high glucose-induced NTDs. In this study, we investigated the effect of NOX4 on the embryonic neuroepithelium in NTDs and reveal a new regulatory mechanism for miR-322 that disrupts neurulation by ameliorating cell apoptosis. METHODS All-trans-retinoic acid (ATRA)-induced mouse model was utilized to study NTDs. RNA pull-down and dual-luciferase reporter assays were used to confirm the interaction between NOX4 and miR-322. In mouse neural stem cells and whole-embryo culture, Western blot and TUNEL were carried out to investigate the effects of miR-322 and NOX4 on neuroepithelium apoptosis in NTD formation. RESULTS NOX4, as a novel target of miR-322, was upregulated in ATRA-induced mouse model of NTDs. In mouse neural stem cells, the expression of NOX4 was inhibited by miR-322; still further, NOX4-triggered apoptosis was also suppressed by miR-322. Moreover, in whole-embryo culture, injection of the miR-322 mimic into the amniotic cavity attenuated cell apoptosis in NTD formation by silencing NOX4. CONCLUSION miR-322/NOX4 plays a crucial role in apoptosis-induced NTD formation, which may provide a new understanding of the mechanism of embryonic NTDs and a basis for potential therapeutic target against NTDs.
Collapse
Affiliation(s)
- Yu-Si Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Tian-Chu Huang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiao-Wei Wei
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wei Ma
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yi-Wen He
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wen-Ting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Jie-Ting Huang
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Duan Zhao
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Shan-Shan Jia
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Fang Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Yu-Zuo Bai
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wei-Lin Wang
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Zheng-Wei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
22
|
Ding Z, Zhou H, McCauley N, Ko G, Zhang KK, Xie L. In ovo hyperglycemia causes congenital limb defects in chicken embryos via disruption of cell proliferation and apoptosis. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165955. [PMID: 32877749 DOI: 10.1016/j.bbadis.2020.165955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/05/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
Abstract
While the correlation between diabetes during pregnancy and birth defects is well-established, how hyperglycemia causes developmental abnormalities remains unclear. In this study, we developed a novel "hyperglycemic" chicken embryonic model by administrating various doses of glucose to fertilized eggs at embryonic stages HH16 or HH24. When the embryos were collected at HH35, the LD50 was 1.57 g/Kg under HH16 treatment and 0.93 g/Kg under HH24 treatment, indicating that "hyperglycemic" environments can be lethal for the embryos. When exposed to a dose equal to or higher than 1 g/Kg glucose at HH16 or HH24, more than 40% of the surviving chicken embryos displayed heart defects and/or limb defects. The limb defects were associated with proliferation defects of both the wing and leg buds indicated by reduced numbers of p-H3S10 labeled cells. These limb defects were also associated with ectopic apoptosis in the leg bud and expression changes of key apoptotic genes. Furthermore, glucose treatment induced decreased expression of genes involved in Shh-signaling, chondrogenesis, and digit patterning in the limb bud. In summary, our data demonstrated that a high-glucose environment induces congenital heart and limb defects associated with disrupted cell proliferation and apoptosis, possibly through depressed Shh-signaling.
Collapse
Affiliation(s)
- Zehuan Ding
- Department of Nutrition, Texas A&M University, College Station, TX, United States of America
| | - Huijuan Zhou
- Department of Statistics, Texas A&M University, College Station, TX, United States of America
| | - Naomi McCauley
- Department of Nutrition, Texas A&M University, College Station, TX, United States of America
| | - Gladys Ko
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States of America
| | - Ke K Zhang
- Department of Nutrition, Texas A&M University, College Station, TX, United States of America; Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX, United States of America
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX, United States of America.
| |
Collapse
|
23
|
Chen ACH, Lee KF, Yeung WSB, Lee YL. Human embryonic stem cells as an in vitro model for studying developmental origins of type 2 diabetes. World J Stem Cells 2020; 12:761-775. [PMID: 32952857 PMCID: PMC7477660 DOI: 10.4252/wjsc.v12.i8.761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/28/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023] Open
Abstract
The developmental origins of health and diseases (DOHaD) is a concept stating that adverse intrauterine environments contribute to the health risks of offspring. Since the theory emerged more than 30 years ago, many epidemiological and animal studies have confirmed that in utero exposure to environmental insults, including hyperglycemia and chemicals, increased the risk of developing noncommunicable diseases (NCDs). These NCDs include metabolic syndrome, type 2 diabetes, and complications such as diabetic cardiomyopathy. Studying the effects of different environmental insults on early embryo development would aid in understanding the underlying mechanisms by which these insults promote NCD development. Embryonic stem cells (ESCs) have also been utilized by researchers to study the DOHaD. ESCs have pluripotent characteristics and can be differentiated into almost every cell lineage; therefore, they are excellent in vitro models for studying early developmental events. More importantly, human ESCs (hESCs) are the best alternative to human embryos for research because of ethical concerns. In this review, we will discuss different maternal conditions associated with DOHaD, focusing on the complications of maternal diabetes. Next, we will review the differentiation protocols developed to generate different cell lineages from hESCs. Additionally, we will review how hESCs are utilized as a model for research into the DOHaD. The effects of environmental insults on hESC differentiation and the possible involvement of epigenetic regulation will be discussed.
Collapse
Affiliation(s)
- Andy Chun-Hang Chen
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen 518053, Guangdong Province, China
| | - Kai Fai Lee
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen 518053, Guangdong Province, China
| | - William Shu Biu Yeung
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen 518053, Guangdong Province, China
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen 518053, Guangdong Province, China
| |
Collapse
|
24
|
Bitew ZW, Worku T, Alebel A, Alemu A. Magnitude and Associated Factors of Neural Tube Defects in Ethiopia: A Systematic Review and Meta-Analysis. Glob Pediatr Health 2020; 7:2333794X20939423. [PMID: 32743026 PMCID: PMC7376379 DOI: 10.1177/2333794x20939423] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/27/2020] [Accepted: 06/12/2020] [Indexed: 01/30/2023] Open
Abstract
Neural tube defects remain a major problem in developing countries, but there are limited comprehensive national reports to date in Ethiopia. Therefore, this study aimed to assess the prevalence of neural tube defects and associated factors in Ethiopia. Electronic databases and other sources were used to retrieve studies. Fifteen out of 862 studies were included in the final analysis. The estimated pooled prevalence of neural tube defects among children in Ethiopia was 63.3 cases per 10 000 children. The pooled prevalence of spinal bifida, anencephaly, and encephalocele was 41.09, 18.90, and 1.07 per 10 000 children, respectively. Previous family history and unplanned pregnancy were risk factors for neural tube defects. Folic acid supplementation during the first trimester of pregnancy was found to be protective. Neural tube defects are widespread in Ethiopia. Hence, fortification of food with folic acid or folic acid supplementation during childbearing age is recommended.
Collapse
Affiliation(s)
| | | | | | - Ayinalem Alemu
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| |
Collapse
|
25
|
Maternal diet intervention before pregnancy primes offspring lipid metabolism in liver. J Transl Med 2020; 100:553-569. [PMID: 31748681 PMCID: PMC7102928 DOI: 10.1038/s41374-019-0344-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/17/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has a developmental origin and is influenced in utero. We aimed to evaluate if maternal diet intervention before pregnancy would be beneficial to reduce the risk of offspring NAFLD. In our study, female mice were either on a normal-fat diet (NF group), or a high-fat diet for 12 weeks and continued on this diet throughout pregnancy and lactation (HF group), or switched from HF-to-NF diet 1 week (H1N group), or 9 weeks (H9N group) before pregnancy. Compared with the NF offspring, the H1N and HF, but not the H9N offspring, displayed more severe hepatic steatosis and glucose intolerance. More specifically, an abnormal blood lipid panel was seen in the H1N offspring and abnormal hepatic free fatty acid composition was present in both the HF and H1N offspring, while the H9N offspring displayed both at normal levels. These physiological changes were associated with desensitized hepatic insulin/AKT signaling, increased expression of genes and proteins for de novo lipogenesis and cholesterol synthesis, decreased expression of genes and proteins for fatty acid oxidation, increased Pcsk9 expression, and hypoactivation of 5' AMP-activated protein kinase (AMPK) signaling in the HF and H1N offspring. However, these effects were completely or partially rescued in the H9N offspring. In summary, we found that early maternal diet intervention is effective in reducing the risk of offspring NAFLD caused by maternal HF diet. These findings provide significant support to develop effective diet intervention strategies and policies for prevention of obesity and NAFLD to promote optimal health outcomes for mothers and children.
Collapse
|
26
|
Chen X, Shen WB, Yang P, Dong D, Sun W, Yang P. High Glucose Inhibits Neural Stem Cell Differentiation Through Oxidative Stress and Endoplasmic Reticulum Stress. Stem Cells Dev 2019; 27:745-755. [PMID: 29695191 DOI: 10.1089/scd.2017.0203] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Maternal diabetes induces neural tube defects by suppressing neurogenesis in the developing neuroepithelium. Our recent study further revealed that high glucose inhibited embryonic stem cell differentiation into neural lineage cells. However, the mechanism whereby high glucose suppresses neural differentiation is unclear. To investigate whether high glucose-induced oxidative stress and endoplasmic reticulum (ER) stress lead to the inhibition of neural differentiation, the effect of high glucose on neural stem cell (the C17.2 cell line) differentiation was examined. Neural stem cells were cultured in normal glucose (5 mM) or high glucose (25 mM) differentiation medium for 3, 5, and 7 days. High glucose suppressed neural stem cell differentiation by significantly decreasing the expression of the neuron marker Tuj1 and the glial cell marker GFAP and the numbers of Tuj1+ and GFAP+ cells. The antioxidant enzyme superoxide dismutase mimetic Tempol reversed high glucose-decreased Tuj1 and GFAP expression and restored the numbers of neurons and glial cells differentiated from neural stem cells. Hydrogen peroxide treatment imitated the inhibitory effect of high glucose on neural stem cell differentiation. Both high glucose and hydrogen peroxide triggered ER stress, whereas Tempol blocked high glucose-induced ER stress. The ER stress inhibitor, 4-phenylbutyrate, abolished the inhibition of high glucose or hydrogen peroxide on neural stem cell differentiation. Thus, oxidative stress and its resultant ER stress mediate the inhibitory effect of high glucose on neural stem cell differentiation.
Collapse
Affiliation(s)
- Xi Chen
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Wei-Bin Shen
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Penghua Yang
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Daoyin Dong
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland
| | - Winny Sun
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland.,2 Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Peixin Yang
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine , Baltimore, Maryland.,2 Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
27
|
Li Z, Zhu H, Liu C, Wang Y, Wang D, Liu H, Cao W, Hu Y, Lin Q, Tong C, Lu M, Sachinidis A, Li L, Peng L. GSK-3β inhibition protects the rat heart from the lipopolysaccharide-induced inflammation injury via suppressing FOXO3A activity. J Cell Mol Med 2019; 23:7796-7809. [PMID: 31503410 PMCID: PMC6815822 DOI: 10.1111/jcmm.14656] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/11/2019] [Accepted: 08/15/2019] [Indexed: 12/25/2022] Open
Abstract
Sepsis‐induced cardiac dysfunction represents a main cause of death in intensive care units. Previous studies have indicated that GSK‐3β is involved in the modulation of sepsis. However, the signalling details of GSK‐3β regulation in endotoxin lipopolysaccharide (LPS)‐induced septic myocardial dysfunction are still unclear. Here, based on the rat septic myocardial injury model, we found that LPS could induce GSK‐3β phosphorylation at its active site (Y216) and up‐regulate FOXO3A level in primary cardiomyocytes. The FOXO3A expression was significantly reduced by GSK‐3β inhibitors and further reversed through β‐catenin knock‐down. This pharmacological inhibition of GSK‐3β attenuated the LPS‐induced cell injury via mediating β‐catenin signalling, which could be abolished by FOXO3A activation. In vivo, GSK‐3β suppression consistently improved cardiac function and relieved heart injury induced by LPS. In addition, the increase in inflammatory cytokines in LPS‐induced model was also blocked by inhibition of GSK‐3β, which curbed both ERK and NF‐κB pathways, and suppressed cardiomyocyte apoptosis via activating the AMP‐activated protein kinase (AMPK). Our results demonstrate that GSK‐3β inhibition attenuates myocardial injury induced by endotoxin that mediates the activation of FOXO3A, which suggests a potential target for the therapy of septic cardiac dysfunction.
Collapse
Affiliation(s)
- Zhigang Li
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huifang Zhu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chang Liu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yumei Wang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Duo Wang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huan Liu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenze Cao
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Hu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qin Lin
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chang Tong
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Lu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine, Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Li Li
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Luying Peng
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Basu M, Garg V. Maternal hyperglycemia and fetal cardiac development: Clinical impact and underlying mechanisms. Birth Defects Res 2019; 110:1504-1516. [PMID: 30576094 DOI: 10.1002/bdr2.1435] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/16/2018] [Indexed: 12/15/2022]
Abstract
Congenital heart disease (CHD) is the most common type of birth defect and is both a significant pediatric and adult health problem, in light of a growing population of survivors. The etiology of CHD has been considered to be multifactorial with genetic and environmental factors playing important roles. The combination of advances in cardiac developmental biology, which have resulted in the elucidation of molecular pathways regulating normal cardiac morphogenesis, and genome sequencing technology have allowed the discovery of numerous genetic contributors of CHD ranging from chromosomal abnormalities to single gene variants. Conversely, mechanistic details of the contribution of environmental factors to CHD remain unknown. Maternal diabetes mellitus (matDM) is a well-established and increasingly prevalent environmental risk factor for CHD, but the underlying etiologic mechanisms by which pregestational matDM increases the vulnerability of embryos to cardiac malformations remains largely elusive. Here, we will briefly discuss the multifactorial etiology of CHD with a focus on the epidemiologic link between matDM and CHD. We will describe the animal models used to study the underlying mechanisms between matDM and CHD and review the numerous cellular and molecular pathways affected by maternal hyperglycemia in the developing heart. Last, we discuss how this increased understanding may open the door for the development of novel prevention strategies to reduce the incidence of CHD in this high-risk population.
Collapse
Affiliation(s)
- Madhumita Basu
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Vidu Garg
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
29
|
Han L, Jiang Z, Zheng X, Qiu J, Hu Y, Li X. Progress in Development of Interventions to Prevent Birth Defects in Diabetic Pregnancies. Chem Pharm Bull (Tokyo) 2019; 67:648-653. [DOI: 10.1248/cpb.c18-01013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Zhe Jiang
- Yanbian University Hospital
- Yanbian University College of Pharmacy
| | | | - Jun Qiu
- Yanbian University Hospital
- Yanbian University College of Pharmacy
| | - Yawen Hu
- Yanbian University Hospital
- Yanbian University College of Pharmacy
| | - Xuezheng Li
- Yanbian University Hospital
- Yanbian University College of Pharmacy
| |
Collapse
|
30
|
Huang Y, Ren A, Wang L, Jin L, Lin S, Li Z, McDonald JA. Casp8 hypomethylation and neural tube defects in association with polycyclic aromatic hydrocarbon exposure. Clin Epigenetics 2019; 11:72. [PMID: 31064411 PMCID: PMC6505285 DOI: 10.1186/s13148-019-0673-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 04/26/2019] [Indexed: 12/25/2022] Open
Abstract
Background Epidemiological studies have found that prenatal exposure to polycyclic aromatic hydrocarbons (PAHs) is associated with increased risk for neural tube defects (NTDs). Aberrant DNA methylation, excessive apoptosis, and oxidative stress have been implied as the mechanism underlying the association between PAH exposure and NTDs, respectively. However, the role of DNA methylation aberration of apoptotic initiator CASP8 (caspase-8, apoptosis-related cysteine peptidase) in the formation of NTDs in association with PAH exposure is not known. By combining a case–control study and mouse model, we aimed to explore the full spectrum of the links from PAH exposure, oxidative stress, CASP8 methylation change, caspase-8 activation, apoptosis, to NTD formation. Results Hypomethylation of CASP8 promoter was noticed in the microarray profiled by Infinium HumanMethylation450 BeadChip using neural tissues from 10 terminated NTD fetuses and 8 terminated non-malformed fetuses (14 CpG sites, with β difference ranging between 8.8 and 26.3%), and was validated in a larger case–control sample performed with neural tissues from 80 NTD cases and 32 non-malformed fetuses, using the Sequenom MassARRAY system (7 CpG sites). Hypomethylation of CASP8 was a risk factor for NTDs (aOR = 1.11; 95% CI, 1.05–1.17) based on the logistic regression model. According to Pearson’s correlation, methylation levels of CASP8 were inversely correlated with PAH concentrations in maternal serum and with oxidative stress markers in fetal neural tissues (p < 0.05). In the animal study, increased NTD rates (13.5% frequency), Casp8 hypomethylation, caspase-8 upregulation, increased caspase-8 cleavage, and excessive apoptosis were found in mouse embryos cultured with benz(a)pyrene (BaP) in vitro. Antioxidant N-acetyl-L-cysteine (NAC) and BaP co-treatment attenuated the changes found in BaP treatment group. Conclusions Hypomethylation of Casp8 promoter is associated with the formation of NTDs, and Casp8 hypomethylation may be induced by oxidative stress that resulted from exposure to PAHs. Electronic supplementary material The online version of this article (10.1186/s13148-019-0673-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yun Huang
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, 100191, China.,Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Aiguo Ren
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, 100191, China. .,Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China.
| | - Linlin Wang
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, 100191, China. .,Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China.
| | - Lei Jin
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, 100191, China.,Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Shanshan Lin
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, 100191, China.,Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Zhiwen Li
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, 100191, China.,Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Jasmine A McDonald
- Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th Street, New York, NY, 10032, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, 10032, USA
| |
Collapse
|
31
|
Yang P, Xu C, Reece EA, Chen X, Zhong J, Zhan M, Stumpo DJ, Blackshear PJ, Yang P. Tip60- and sirtuin 2-regulated MARCKS acetylation and phosphorylation are required for diabetic embryopathy. Nat Commun 2019; 10:282. [PMID: 30655546 PMCID: PMC6336777 DOI: 10.1038/s41467-018-08268-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/21/2018] [Indexed: 12/13/2022] Open
Abstract
Failure of neural tube closure results in severe birth defects and can be induced by high glucose levels resulting from maternal diabetes. MARCKS is required for neural tube closure, but the regulation and of its biological activity and function have remained elusive. Here, we show that high maternal glucose induced MARCKS acetylation at lysine 165 by the acetyltransferase Tip60, which is a prerequisite for its phosphorylation, whereas Sirtuin 2 (SIRT2) deacetylated MARCKS. Phosphorylated MARCKS dissociates from organelles, leading to mitochondrial abnormalities and endoplasmic reticulum stress. Phosphorylation dead MARCKS (PD-MARCKS) reversed maternal diabetes-induced cellular organelle stress, apoptosis and delayed neurogenesis in the neuroepithelium and ameliorated neural tube defects. Restoring SIRT2 expression in the developing neuroepithelium exerted identical effects as those of PD-MARCKS. Our studies reveal a new regulatory mechanism for MARCKS acetylation and phosphorylation that disrupts neurulation under diabetic conditions by diminishing the cellular organelle protective effect of MARCKS.
Collapse
Affiliation(s)
- Penghua Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
| | - Cheng Xu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
| | - E Albert Reece
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, MD, USA.,Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
| | - Xi Chen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
| | - Jianxiang Zhong
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
| | - Min Zhan
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, 21201, MD, USA
| | - Deborah J Stumpo
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA.,Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, NC, 27710, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, MD, USA. .,Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, 21201, MD, USA.
| |
Collapse
|
32
|
Xu C, Chen X, Reece EA, Lu W, Yang P. The increased activity of a transcription factor inhibits autophagy in diabetic embryopathy. Am J Obstet Gynecol 2019; 220:108.e1-108.e12. [PMID: 30312583 DOI: 10.1016/j.ajog.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Maternal diabetes induces neural tube defects and stimulates the activity of the forkhead box O3 (Fox)O3a in the embryonic neuroepithelium. We previously demonstrated that deleting the FOXO3a gene ameliorates maternal diabetes-induced neural tube defects. Macroautophagy (hereafter referred to as "autophagy") is essential for neurulation. Rescuing autophagy suppressed by maternal diabetes in the developing neuroepithelium inhibits neural tube defect formation in diabetic pregnancy. This evidence suggests a possible link between FoxO3a and impaired autophagy in diabetic embryopathy. OBJECTIVE We aimed to determine whether maternal diabetes suppresses autophagy through FoxO3a, and if the transcriptional activity of FoxO3a is required for the induction of diabetic embryopathy. STUDY DESIGN We used a well-established type 1 diabetic embryopathy mouse model, in which diabetes was induced by streptozotocin, for our in vivo studies. To determine if FoxO3a mediates the inhibitory effect of maternal diabetes on autophagy in the developing neuroepithelium, we induced diabetic embryopathy in FOXO3a gene knockout mice and FoxO3a dominant negative transgenic mice. Embryos were harvested at embryonic day 8.5 to determine FoxO3a and autophagy activity and at embryonic day 10.5 for the presence of neural tube defects. We also examined the expression of autophagy-related genes. C17.2 neural stem cells were used for in vitro examination of the potential effects of FoxO3a on autophagy. RESULTS Deletion of the FOXO3a gene restored the autophagy markers, lipidation of microtubule-associated protein 1A/1B-light chain 3I to light chain 3II, in neurulation stage embryos. Maternal diabetes decreased light chain 3I-positive puncta number in the neuroepithelium, which was restored by deleting FoxO3a. Maternal diabetes also decreased the expression of positive regulators of autophagy (Unc-51 like autophagy activating kinase 1, Coiled-coil myosin-like BCL2-interacting protein, and autophagy-related gene 5) and the negative regulator of autophagy, p62. FOXO3a gene deletion abrogated the dysregulation of autophagy genes. In vitro data showed that the constitutively active form of FoxO3a mimicked high glucose in repressing autophagy. In cells cultured under high-glucose conditions, overexpression of the dominant negative FoxO3a mutant blocked autophagy impairment. Dominant negative FoxO3a overexpression in the developing neuroepithelium restored autophagy and significantly reduced maternal diabetes-induced apoptosis and neural tube defects. CONCLUSION Our study revealed that diabetes-induced FoxO3a activation inhibited autophagy in the embryonic neuroepithelium. We also observed that FoxO3a transcriptional activity mediated the teratogenic effect of maternal diabetes because dominant negative FoxO3a prevents maternal diabetes-induced autophagy impairment and neural tube defect formation. Our findings suggest that autophagy activators could be therapeutically effective in treating maternal diabetes-induced neural tube defects.
Collapse
|
33
|
Engineer A, Saiyin T, Lu X, Kucey AS, Urquhart BL, Drysdale TA, Norozi K, Feng Q. Sapropterin Treatment Prevents Congenital Heart Defects Induced by Pregestational Diabetes Mellitus in Mice. J Am Heart Assoc 2018; 7:e009624. [PMID: 30608180 PMCID: PMC6404194 DOI: 10.1161/jaha.118.009624] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/17/2018] [Indexed: 01/05/2023]
Abstract
Background Tetrahydrobiopterin is a cofactor of endothelial NO synthase ( eNOS ), which is critical to embryonic heart development. We aimed to study the effects of sapropterin (Kuvan), an orally active synthetic form of tetrahydrobiopterin on eNOS uncoupling and congenital heart defects ( CHD s) induced by pregestational diabetes mellitus in mice. Methods and Results Adult female mice were induced to pregestational diabetes mellitus by streptozotocin and bred with normal male mice to produce offspring. Pregnant mice were treated with sapropterin or vehicle during gestation. CHD s were identified by histological analysis. Cell proliferation, eNOS dimerization, and reactive oxygen species production were assessed in the fetal heart. Pregestational diabetes mellitus results in a spectrum of CHD s in their offspring. Oral treatment with sapropterin in the diabetic dams significantly decreased the incidence of CHD s from 59% to 27%, and major abnormalities, such as atrioventricular septal defect and double-outlet right ventricle, were absent in the sapropterin-treated group. Lineage tracing reveals that pregestational diabetes mellitus results in decreased commitment of second heart field progenitors to the outflow tract, endocardial cushions, and ventricular myocardium of the fetal heart. Notably, decreased cell proliferation and cardiac transcription factor expression induced by maternal diabetes mellitus were normalized with sapropterin treatment. Furthermore, sapropterin administration in the diabetic dams increased eNOS dimerization and lowered reactive oxygen species levels in the fetal heart. Conclusions Sapropterin treatment in the diabetic mothers improves eNOS coupling, increases cell proliferation, and prevents the development of CHD s in the offspring. Thus, sapropterin may have therapeutic potential in preventing CHD s in pregestational diabetes mellitus.
Collapse
Affiliation(s)
- Anish Engineer
- Department of Physiology and PharmacologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Tana Saiyin
- Department of Physiology and PharmacologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Xiangru Lu
- Department of Physiology and PharmacologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Andrew S. Kucey
- Department of Physiology and PharmacologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Brad L. Urquhart
- Department of Physiology and PharmacologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Thomas A. Drysdale
- Department of Physiology and PharmacologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of PediatricsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Children's Health Research InstituteLondonOntarioCanada
| | - Kambiz Norozi
- Department of PediatricsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Children's Health Research InstituteLondonOntarioCanada
- Department of Paediatric Cardiology and Intensive Care MedicineHannover Medical SchoolHannoverGermany
- Department of Paediatric Cardiology and Intensive Care MedicineUniversity of GöttingenGermany
| | - Qingping Feng
- Department of Physiology and PharmacologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of MedicineSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Children's Health Research InstituteLondonOntarioCanada
| |
Collapse
|
34
|
Ornoy A, Koren G, Yanai J. Is post exposure prevention of teratogenic damage possible: Studies on diabetes, valproic acid, alcohol and anti folates in pregnancy: Animal studies with reflection to human. Reprod Toxicol 2018; 80:92-104. [DOI: 10.1016/j.reprotox.2018.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/06/2018] [Accepted: 05/25/2018] [Indexed: 12/20/2022]
|
35
|
Miyazawa H, Yamamoto M, Yamaguchi Y, Miura M. Mammalian embryos show metabolic plasticity toward the surrounding environment during neural tube closure. Genes Cells 2018; 23:794-802. [PMID: 30088697 DOI: 10.1111/gtc.12626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/17/2018] [Accepted: 06/26/2018] [Indexed: 01/16/2023]
Abstract
Developing embryos rewire energy metabolism for developmental processes. However, little is known about how metabolic rewiring is coupled with development in a spatiotemporal manner. Here, we show that mammalian embryos display plasticity of glucose metabolism in response to the extracellular environment at the neural tube closure (NTC) stage, when the intrauterine environment changes upon placentation. To study how embryos modulate their metabolic state upon environmental change, we analyzed the steady-state level of ATP upon exposure to extrauterine environments using both an enzymatic assay and a genetically encoded ATP sensor. Upon environmental changes, NTC-stage embryos exhibited increased ATP content, whereas embryos before and after NTC did not. The increased ATP in the NTC-stage embryos seemed to depend on glycolysis. Intriguingly, an increase in mitochondrial membrane potential (ΔΨm) was also observed in the neural ectoderm (NE) and the neural plate border of the non-neural ectoderm (NNE) region. This implies that glycolysis can be coupled with the TCA cycle in the NE and the neural plate border depending on environmental context. Disrupting ΔΨm inhibited folding of the cranial neural plate. Thus, we propose that embryos tune metabolic plasticity to enable coupling of glucose metabolism with the extracellular environment at the NTC stage.
Collapse
Affiliation(s)
- Hidenobu Miyazawa
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Masamichi Yamamoto
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Tokyo, Japan.,Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshifumi Yamaguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Hibernation Metabolism, Physiology and Development Group, Institute of Low Temperature Science, Hokkaido University, Sapporo, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
36
|
Chen X, Zhong J, Dong D, Liu G, Yang P. Endoplasmic Reticulum Stress-Induced CHOP Inhibits PGC-1α and Causes Mitochondrial Dysfunction in Diabetic Embryopathy. Toxicol Sci 2018; 158:275-285. [PMID: 28482072 DOI: 10.1093/toxsci/kfx096] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress has been implicated in the development of maternal diabetes-induced neural tube defects (NTDs). ER stress-induced C/EBP homologous protein (CHOP) plays an important role in the pro-apoptotic execution pathways. However, the molecular mechanism underlying ER stress- and CHOP-induced neuroepithelium cell apoptosis in diabetic embryopathy is still unclear. Deletion of the Chop gene significantly reduced maternal diabetes-induced NTDs. CHOP deficiency abrogated maternal diabetes-induced mitochondrial dysfunction and neuroepithelium cell apoptosis. Further analysis demonstrated that CHOP repressed the expression of peroxisome-proliferator-activated receptor-γ coactivator-1α (PGC-1α), an essential regulator for mitochondrial biogenesis and function. Both CHOP deficiency in vivo and knockdown in vitro restore high glucose-suppressed PGC-1α expression. In contrast, CHOP overexpression mimicked inhibition of PGC-1α by high glucose. In response to the ER stress inducer tunicamycin, PGC-1α expression was decreased, whereas the ER stress inhibitor 4-phenylbutyric acid blocked high glucose-suppressed PGC-1α expression. Moreover, maternal diabetes in vivo and high glucose in vitro promoted the interaction between CHOP and the PGC-1α transcriptional regulator CCAAT/enhancer binding protein-β (C/EBPβ), and reduced C/EBPβ binding to the PGC-1α promoter leading to markedly decrease in PGC-1α expression. Together, our findings support the hypothesis that maternal diabetes-induced ER stress increases CHOP expression which represses PGC-1α through suppressing the C/EBPβ transcriptional activity, subsequently induces mitochondrial dysfunction and ultimately results in NTDs.
Collapse
Affiliation(s)
- Xi Chen
- Center for Translational Research, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, People's Republic of China.,Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Jianxiang Zhong
- Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Daoyin Dong
- Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Gentao Liu
- Center for Translational Research, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, People's Republic of China
| | - Peixin Yang
- Center for Translational Research, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, People's Republic of China.,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
37
|
Mazumdar M. Does arsenic increase the risk of neural tube defects among a highly exposed population? A new case-control study in Bangladesh. Birth Defects Res 2018; 109:92-98. [PMID: 27801974 DOI: 10.1002/bdra.23577] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 08/01/2016] [Accepted: 08/12/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Neural tube defects are debilitating birth defects that occur when the developing neural plate fails to close in early gestation. Arsenic induces neural tube defects in animal models, but whether environmental arsenic exposure increases risk of neural tube defects in humans is unknown. METHODS We describe a new case-control study in Bangladesh, a country currently experiencing an epidemic of arsenic poisoning through contaminated drinking water. We plan to understand how arsenic influences risk of neural tube defects in humans through mechanisms that include disruption of maternal glucose and folate metabolism, as well as epigenetic effects. We also investigate whether sweat chloride concentration, a potential new biomarker for arsenic toxicity, can be used to identify women at higher risk for having a child affected by neural tube defect. We will collect dural tissue from cases, obtained at the time of surgical closure of the defect, and believe investigation of these samples will provide insight into the epigenetic mechanisms by which prenatal arsenic exposure affects the developing nervous system. CONCLUSION These studies explore mechanisms by which arsenic may increase risk of neural tube defects in humans and use a unique population with high arsenic exposure to test hypotheses. If successful, these studies may assist countries with high arsenic exposure such as Bangladesh to identify populations at high risk of neural tube defects, as well as direct development of novel screening strategies for maternal risk.Birth Defects Research 109:92-98, 2017.© 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Maitreyi Mazumdar
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Harvard Medical School, Boston, Massachusetts.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
38
|
Lin S, Ren A, Wang L, Huang Y, Wang Y, Wang C, Greene ND. Oxidative Stress and Apoptosis in Benzo[a]pyrene-Induced Neural Tube Defects. Free Radic Biol Med 2018; 116:149-158. [PMID: 29309894 PMCID: PMC5821680 DOI: 10.1016/j.freeradbiomed.2018.01.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022]
Abstract
Neural tube defects (NTDs) are among the most common and severe congenital malformations and result from incomplete closure of the neural tube during early development. Maternal exposure to polycyclic aromatic hydrocarbons (PAHs) has been suggested to be a risk factor for NTDs and previous studies imply that the mechanism underlying the association between PAH exposure and NTDs may involve oxidative stress and apoptosis. The objectives of this study were to investigate whether there is a direct effect of maternal benzo[α] pyrene (BaP) exposure on the closure of the neural tube in mice, and to examine the underlying mechanisms by combining animal experiments and human subject studies. We found that intraperitoneal injection of BaP from embryonic day 7 at a dose of 250 mg kg-1 induced NTDs (13.3% frequency) in ICR mice. BaP exposure significantly increased expression of genes associated with oxidative stress, Cyp1a1, Sod1 and Sod2, while repressing Gpx1. Elevated apoptosis and higher protein expression of cleaved caspase-3 in the neuroepithelium of treated embryos were observed. Pre-treatment with vitamin E, added to food, significantly protected against BaP-induced NTDs (1.4% frequency) (P < 0.05). Vitamin E also partly normalized oxidative stress related gene expression and excess apoptosis in BaP-treated embryos. Examination of human neural tissues revealed that increased levels of protein carbonyl and apoptosis were related with maternal exposure to PAHs and the risk of NTDs. Collectively, these results suggest that BaP exposure could induce NTDs and that this may involve increased oxidative stress and apoptosis, while vitamin E may have a protective effect.
Collapse
Affiliation(s)
- Shanshan Lin
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Center, Peking University, Beijing, China
| | - Aiguo Ren
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Center, Peking University, Beijing, China.
| | - Linlin Wang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Center, Peking University, Beijing, China.
| | - Yun Huang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Center, Peking University, Beijing, China
| | - Yuanyuan Wang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Center, Peking University, Beijing, China
| | - Caiyun Wang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Center, Peking University, Beijing, China
| | - Nicholas D Greene
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, WC1N 1EH, London, United Kingdom
| |
Collapse
|
39
|
Zhao Y, Dong D, Reece EA, Wang AR, Yang P. Oxidative stress-induced miR-27a targets the redox gene nuclear factor erythroid 2-related factor 2 in diabetic embryopathy. Am J Obstet Gynecol 2018; 218:136.e1-136.e10. [PMID: 29100869 DOI: 10.1016/j.ajog.2017.10.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/23/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Maternal diabetes induces neural tube defects, and oxidative stress is a causal factor for maternal diabetes-induced neural tube defects. The redox gene nuclear factor erythroid 2-related factor 2 is the master regulator of the cellular antioxidant system. OBJECTIVE In this study, we aimed to determine whether maternal diabetes inhibits nuclear factor erythroid 2-related factor 2 expression and nuclear factor erythroid 2-related factor 2-controlled antioxidant genes through the redox-sensitive miR-27a. STUDY DESIGN We used a well-established type 1 diabetic embryopathy mouse model induced by streptozotocin for our in vivo studies. Embryos at embryonic day 8.5 were harvested for analysis of nuclear factor erythroid 2-related factor 2, nuclear factor erythroid 2-related factor 2-controlled antioxidant genes, and miR-27a expression. To determine if mitigating oxidative stress inhibits the increase of miR-27a and the decrease of nuclear factor erythroid 2-related factor 2 expression, we induced diabetic embryopathy in superoxide dismutase 2 (mitochondrial-associated antioxidant gene)-overexpressing mice. This model exhibits reduced mitochondria reactive oxygen species even in the presence of hyperglycemia. To investigate the causal relationship between miR-27a and nuclear factor erythroid 2-related factor 2 in vitro, we examined C17.2 neural stem cells under normal and high-glucose conditions. RESULTS We observed that the messenger RNA and protein levels of nuclear factor erythroid 2-related factor 2 were significantly decreased in embryos on embryonic day 8.5 from diabetic dams compared to those from nondiabetic dams. High-glucose also significantly decreased nuclear factor erythroid 2-related factor 2 expression in a dose- and time-dependent manner in cultured neural stem cells. Our data revealed that miR-27a was up-regulated in embryos on embryonic day 8.5 exposed to diabetes, and that high glucose increased miR-27a levels in a dose- and time-dependent manner in cultured neural stem cells. In addition, we found that a miR-27a inhibitor abrogated the inhibitory effect of high glucose on nuclear factor erythroid 2-related factor 2 expression, and a miR-27a mimic suppressed nuclear factor erythroid 2-related factor 2 expression in cultured neural stem cells. Furthermore, our data indicated that the nuclear factor erythroid 2-related factor 2-controlled antioxidant enzymes glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione S-transferase A1 were down-regulated by maternal diabetes in embryos on embryonic day 8.5 and high glucose in cultured neural stem cells. Inhibiting miR-27a restored expression of glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione S-transferase A1. Overexpressing superoxide dismutase 2 reversed the maternal diabetes-induced increase of miR-27a and suppression of nuclear factor erythroid 2-related factor 2 and nuclear factor erythroid 2-related factor 2-controlled antioxidant enzymes. CONCLUSION Our study demonstrates that maternal diabetes-induced oxidative stress increases miR-27a, which, in turn, suppresses nuclear factor erythroid 2-related factor 2 and its responsive antioxidant enzymes, resulting in diabetic embryopathy.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Daoyin Dong
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - E Albert Reece
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Ashley R Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Peixin Yang
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Chashan University-town, Wenzhou, Zhejiang, China; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
40
|
Demir N, Başaranoğlu M, Huyut Z, Değer İ, Karaman K, Şekeroğlu MR, Tuncer O. The relationship between mother and infant plasma trace element and heavy metal levels and the risk of neural tube defect in infants. J Matern Fetal Neonatal Med 2017; 32:1433-1440. [DOI: 10.1080/14767058.2017.1408064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Nihat Demir
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Murat Başaranoğlu
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Zübeyir Huyut
- Department of Medical Biochemistry, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - İbrahim Değer
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Kamuran Karaman
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - M. Ramazan Şekeroğlu
- Department of Medical Biochemistry, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Oğuz Tuncer
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| |
Collapse
|
41
|
Miyazawa H, Yamaguchi Y, Sugiura Y, Honda K, Kondo K, Matsuda F, Yamamoto T, Suematsu M, Miura M. Rewiring of embryonic glucose metabolism via suppression of PFK-1 and aldolase during mouse chorioallantoic branching. Development 2017; 144:63-73. [PMID: 28049690 PMCID: PMC5278628 DOI: 10.1242/dev.138545] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 11/19/2016] [Indexed: 12/31/2022]
Abstract
Adapting the energy metabolism state to changing bioenergetic demands is essential for mammalian development accompanying massive cell proliferation and cell differentiation. However, it remains unclear how developing embryos meet the changing bioenergetic demands during the chorioallantoic branching (CB) stage, when the maternal-fetal exchange of gases and nutrients is promoted. In this study, using metabolome analysis with mass-labeled glucose, we found that developing embryos redirected glucose carbon flow into the pentose phosphate pathway via suppression of the key glycolytic enzymes PFK-1 and aldolase during CB. Concomitantly, embryos exhibited an increase in lactate pool size and in the fractional contribution of glycolysis to lactate biosynthesis. Imaging mass spectrometry visualized lactate-rich tissues, such as the dorsal or posterior neural tube, somites and head mesenchyme. Furthermore, we found that the heterochronic gene Lin28a could act as a regulator of the metabolic changes observed during CB. Perturbation of glucose metabolism rewiring by suppressing Lin28a downregulation resulted in perinatal lethality. Thus, our work demonstrates that developing embryos rewire glucose metabolism following CB for normal development. Highlighted article: Metabolic remodelling during E8.5 to E10.5 in mouse redirects glucose carbon into the pentose phosphate pathway, a process partially regulated by Lin28a.
Collapse
Affiliation(s)
- Hidenobu Miyazawa
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshifumi Yamaguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan .,Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0075, Japan
| | - Yuki Sugiura
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0075, Japan.,Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kurara Honda
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Koki Kondo
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Fumio Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takehiro Yamamoto
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan .,Agency for Medical Research and Development-Core Research for Evolutional Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-1004, Japan
| |
Collapse
|
42
|
Yamaguchi Y, Miyazawa H, Miura M. Neural tube closure and embryonic metabolism. Congenit Anom (Kyoto) 2017; 57:134-137. [PMID: 28295633 DOI: 10.1111/cga.12219] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/04/2017] [Accepted: 03/09/2017] [Indexed: 12/15/2022]
Abstract
Neural tube closure (NTC) is an embryonic process during formation of the mammalian central nervous system. Disruption of the dynamic, sequential events of NTC can cause neural tube defects (NTD) leading to spina bifida and anencephaly in the newborn. NTC is affected by inherent factors such as genetic mutation or if the mother is exposed to certain environmental factors such as intake of harmful chemicals, maternal infection, irradiation, malnutrition, and inadequate or excessive intake of specific nutrients. Although effects of these stress factors on NTC have been intensively studied, the metabolic state of a normally developing embryo remains unclear. State-of-the art mass spectrometry techniques have enabled detailed study of embryonic metabolite profiles and their distribution within tissues. This approach has demonstrated that glucose metabolism is altered during NTC stages involving chorioallantoic branching. An understanding of embryonic metabolic rewiring would help reveal the etiology of NTD caused by environmental factors.
Collapse
Affiliation(s)
- Yoshifumi Yamaguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Agency for Medical Research and Development-Core Research for Evolutional Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Hidenobu Miyazawa
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Agency for Medical Research and Development-Core Research for Evolutional Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
43
|
Wang L, Lin S, Yi D, Huang Y, Wang C, Jin L, Liu J, Zhang Y, Ren A. Apoptosis, Expression of PAX3 and P53, and Caspase Signal in Fetuses with Neural Tube Defects. Birth Defects Res 2017; 109:1596-1604. [PMID: 28786179 DOI: 10.1002/bdr2.1094] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/08/2017] [Accepted: 07/03/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Neural tube defects (NTDs) are among the most common and severe congenital malformations of the central nervous system. Animal studies have shown that apoptosis is involved in the development of NTDs. However, little evidence is available from human studies. We aim to examine the level of apoptosis and expression of apoptosis-regulating proteins of human terminated fetuses. METHODS A total of 37 NTD cases and 21 controls from pregnancy terminations were recruited. Tissues of the central nervous system were obtained through autopsy. Apoptosis of neuroepithelial cells was examined by terminal deoxynucleotidyl transferase-mediated deoxyuridinetriphosphate nick end-labeling (TUNEL) assay. Expression of PAX3, p53, and caspase 3/8/9 in central nervous tissue was measured using Western blotting. RESULTS More TUNEL-positive apoptosis cells were observed in the central nervous tissue of NTD cases than those of controls (p < 0.05). In spinal cord tissue, lower PAX3 expression, higher p53 expression, and increased levels of cleaved caspase 3(17kD) and cleaved caspase 8 (18kD) were found in anencephaly cases but not in spina bifida cases when compared with controls. In brain tissue, levels of PAX3 were significantly reduced in both encephalocele and spina bifida subtypes; the expression levels of cleaved caspase 3(17 kD) of encephalocele cases and cleaved caspase 8(47/45 kD) in spina bifida cases were higher than in controls; no difference was found in the expression of p53 or caspase 9 between NTDs and controls. CONCLUSION These findings provide some evidence that excessive apoptosis in fetal central nervous tissues may be associated with the development of human NTDs. Birth Defects Research 109:1596-1604, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Linlin Wang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Shanshan Lin
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Deqing Yi
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Yun Huang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Caiyun Wang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Lei Jin
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Jufen Liu
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Yali Zhang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| | - Aiguo Ren
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, and Department of Epidemiology and Health Statistics, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
44
|
Wang F, Xu C, Reece EA, Li X, Wu Y, Harman C, Yu J, Dong D, Wang C, Yang P, Zhong J, Yang P. Protein kinase C-alpha suppresses autophagy and induces neural tube defects via miR-129-2 in diabetic pregnancy. Nat Commun 2017; 8:15182. [PMID: 28474670 PMCID: PMC5424165 DOI: 10.1038/ncomms15182] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 03/03/2017] [Indexed: 12/19/2022] Open
Abstract
Gene deletion-induced autophagy deficiency leads to neural tube defects (NTDs), similar to those in diabetic pregnancy. Here we report the key autophagy regulators modulated by diabetes in the murine developing neuroepithelium. Diabetes predominantly leads to exencephaly, induces neuroepithelial cell apoptosis and suppresses autophagy in the forebrain and midbrain of NTD embryos. Deleting the Prkca gene, which encodes PKCα, reverses diabetes-induced autophagy impairment, cellular organelle stress and apoptosis, leading to an NTD reduction. PKCα increases the expression of miR-129-2, which is a negative regulator of autophagy. miR-129-2 represses autophagy by directly targeting PGC-1α, a positive regulator for mitochondrial function, which is disturbed by maternal diabetes. PGC-1α supports neurulation by stimulating autophagy in neuroepithelial cells. These findings identify two negative autophagy regulators, PKCα and miR-129-2, which mediate the teratogenicity of hyperglycaemia leading to NTDs. We also reveal a function for PGC-1α in embryonic development through promoting autophagy and ameliorating hyperglycaemia-induced NTDs.
Collapse
Affiliation(s)
- Fang Wang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Cheng Xu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - E. Albert Reece
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Xuezheng Li
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Yanqing Wu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Christopher Harman
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Jingwen Yu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Daoyin Dong
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Cheng Wang
- Department of Obstetrics, Gynecology, Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Penghua Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Jianxiang Zhong
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| |
Collapse
|
45
|
Yang P, Yang WW, Chen X, Kaushal S, Dong D, Shen WB. Maternal diabetes and high glucose in vitro trigger Sca1 + cardiac progenitor cell apoptosis through FoxO3a. Biochem Biophys Res Commun 2016; 482:575-581. [PMID: 27856257 DOI: 10.1016/j.bbrc.2016.11.076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/12/2016] [Indexed: 12/21/2022]
Abstract
Recent controversies surrounding the authenticity of c-kit+ cardiac progenitor cells significantly push back the advance in regenerative therapies for cardiovascular diseases. There is an urgent need for research in characterizing alternative types of cardiac progenitor cells. Towards this goal, in the present study, we determined the effect of maternal diabetes on Sca1+ cardiac progenitor cells. Maternal diabetes induced caspase 3-dependent apoptosis in Sca1+ cardiac progenitor cells derived from embryonic day 17.5 (E17.5). Similarly, high glucose in vitro but not the glucose osmotic control mannitol triggered Sca1+ cardiac progenitor cell apoptosis in a dose- and time-dependent manner. Both maternal diabetes and high glucose in vitro activated the pro-apoptotic transcription factor, Forkhead O 3a (FoxO3a) via dephosphorylation at threonine 32 (Thr-32) residue. foxo3a gene deletion abolished maternal diabetes-induced Sca1+ cardiac progenitor cell apoptosis. The dominant negative FoxO3a mutant without the transactivation domain from the C terminus blocked high glucose-induced Sca1+ cardiac progenitor cell apoptosis, whereas the constitutively active FoxO3a mutant with the three phosphorylation sites, Thr-32, Ser-253, and Ser-315, being replaced by alanine residues mimicked the pro-apoptotic effect of high glucose. Thus, maternal diabetes and high glucose in vitro may limit the regenerative potential of Sca1+ cardiac progenitor cells by inducing apoptosis through FoxO3a activation. These findings will serve as the guide in optimizing the autologous therapy using Sca1+ cardiac progenitor cells in cardiac defect babies born exposed to maternal diabetes.
Collapse
Affiliation(s)
- Penghua Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wendy W Yang
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xi Chen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sunjay Kaushal
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Daoyin Dong
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
46
|
Zhong J, Xu C, Reece EA, Yang P. The green tea polyphenol EGCG alleviates maternal diabetes-induced neural tube defects by inhibiting DNA hypermethylation. Am J Obstet Gynecol 2016; 215:368.e1-368.e10. [PMID: 26979632 PMCID: PMC5270539 DOI: 10.1016/j.ajog.2016.03.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Maternal diabetes increases the risk of neural tube defects in offspring. Our previous study demonstrated that the green tea polyphenol, Epigallocatechin gallate, inhibits high glucose-induced neural tube defects in cultured embryos. However, the therapeutic effect of Epigallocatechin gallate on maternal diabetes-induced neural tube defects is still unclear. OBJECTIVE We aimed to examine whether Epigallocatechin gallate treatment can reduce maternal diabetes-induced DNA methylation and neural tube defects. STUDY DESIGN Nondiabetic and diabetic pregnant mice at embryonic day 5.5 were given drinking water with or without 1 or 10 μM Epigallocatechin gallate. At embryonic day 8.75, embryos were dissected from the visceral yolk sac for the measurement of the levels and activity of DNA methyltransferases, the levels of global DNA methylation, and methylation in the CpG islands of neural tube closure essential gene promoters. embryonic day 10.5 embryos were examined for neural tube defect incidence. RESULTS Epigallocatechin gallate treatment did not affect embryonic development because embryos from nondiabetic dams treated with Epigallocatechin gallate did not exhibit any neural tube defects. Treatment with 1 μM Epigallocatechin gallate did not reduce maternal diabetes-induced neural tube defects significantly. Embryos from diabetic dams treated with 10 μM Epigallocatechin gallate had a significantly lower neural tube defect incidence compared with that of embryos without Epigallocatechin gallate treatment. Epigallocatechin gallate reduced neural tube defect rates from 29.5% to 2%, an incidence that is comparable with that of embryos from nondiabetic dams. Ten micromoles of Epigallocatechin gallate treatment blocked maternal diabetes-increased DNA methyltransferases 3a and 3b expression and their activities, leading to the suppression of global DNA hypermethylation. Additionally, 10 μM Epigallocatechin gallate abrogated maternal diabetes-increased DNA methylation in the CpG islands of neural tube closure essential genes, including Grhl3, Pax3, and Tulp3. CONCLUSION Epigallocatechin gallate reduces maternal diabetes-induced neural tube defects formation and blocks the enhanced expression and activity of DNA methyltransferases, leading to the suppression of DNA hypermethylation and the restoration of neural tube closure essential gene expression. These observations suggest that Epigallocatechin gallate supplements could mitigate the teratogenic effects of hyperglycemia on the developing embryo and prevent diabetes-induced neural tube defects.
Collapse
Affiliation(s)
- Jianxiang Zhong
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Cheng Xu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - E Albert Reece
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Peixin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
47
|
Type 2 diabetes mellitus induces congenital heart defects in murine embryos by increasing oxidative stress, endoplasmic reticulum stress, and apoptosis. Am J Obstet Gynecol 2016; 215:366.e1-366.e10. [PMID: 27038779 DOI: 10.1016/j.ajog.2016.03.036] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Maternal type 1 and 2 diabetes mellitus are strongly associated with high rates of severe structural birth defects, including congenital heart defects. Studies in type 1 diabetic embryopathy animal models have demonstrated that cellular stress-induced apoptosis mediates the teratogenicity of maternal diabetes leading to congenital heart defect formation. However, the mechanisms underlying maternal type 2 diabetes mellitus-induced congenital heart defects remain largely unknown. OBJECTIVE We aim to determine whether oxidative stress, endoplasmic reticulum stress, and excessive apoptosis are the intracellular molecular mechanisms underlying maternal type 2 diabetes mellitus-induced congenital heart defects. STUDY DESIGN A mouse model of maternal type 2 diabetes mellitus was established by feeding female mice a high-fat diet (60% fat). After 15 weeks on the high-fat diet, the mice showed characteristics of maternal type 2 diabetes mellitus. Control dams were either fed a normal diet (10% fat) or the high-fat diet during pregnancy only. Female mice from the high-fat diet group and the 2 control groups were mated with male mice that were fed a normal diet. At E12.5, embryonic hearts were harvested to determine the levels of lipid peroxides and superoxide, endoplasmic reticulum stress markers, cleaved caspase 3 and 8, and apoptosis. E17.5 embryonic hearts were harvested for the detection of congenital heart defect formation using India ink vessel patterning and histological examination. RESULTS Maternal type 2 diabetes mellitus significantly induced ventricular septal defects and persistent truncus arteriosus in the developing heart, along with increasing oxidative stress markers, including superoxide and lipid peroxidation; endoplasmic reticulum stress markers, including protein levels of phosphorylated-protein kinase RNA-like endoplasmic reticulum kinase, phosphorylated-IRE1α, phosphorylated-eIF2α, C/EBP homologous protein, and binding immunoglobulin protein; endoplasmic reticulum chaperone gene expression; and XBP1 messenger RNA splicing, as well as increased cleaved caspase 3 and 8 in embryonic hearts. Furthermore, maternal type 2 diabetes mellitus triggered excessive apoptosis in ventricular myocardium, endocardial cushion, and outflow tract of the embryonic heart. CONCLUSION Similar to those observations in type 1 diabetic embryopathy, maternal type 2 diabetes mellitus causes heart defects in the developing embryo manifested with oxidative stress, endoplasmic reticulum stress, and excessive apoptosis in heart cells.
Collapse
|
48
|
Wu Y, Zhang Z, Liao X, Qi L, Liu Y, Wang Z. Effect of high-fat diet-induced obesity on the Akt/FoxO/Smad signaling pathway and the follicular development of the mouse ovary. Mol Med Rep 2016; 14:3894-900. [PMID: 27574010 DOI: 10.3892/mmr.2016.5671] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/10/2016] [Indexed: 11/06/2022] Open
Abstract
Obesity has a negative effect on ovarian functions, which is reported to increase the risk of infertility. The mechanism underlying obesity‑induced infertility is not yet clear. The present study established a high‑fat diet (HFD)‑induced obesity mouse model to elucidate the mechanisms underlying the effect of HFD‑induced obesity on follicular development in the mouse ovary. The 4‑week‑old female mice were fed with HFD or normal control (NC) diet for 15 or 20 weeks. Body mass index was used to demonstrate that the mice were obese following HFD treatment. The follicular development of the ovaries from the HFD group mice was retarded in a time‑dependent manner, as demonstrated by morphological and histological examination of the ovaries. Further investigation via western blot analysis demonstrated that the activity of the transcription factor, forkhead box O3a (FoxO3a), was increased by HFD through downregulated FoxO3a phosphorylation, which may contribute to the inhibited development of ovarian follicles. To determine the regulatory mechanism of FoxO3 on the follicular development, the expression levels of FoxO3a target protein, Smad1/5/8, were also determined and there was significant decrease in phosphorylated Smad1/5/8 in the ovaries from the HFD group compared with the NC group, indicating that FoxO3a/Smad1/5/8 may be important in the regulation of follicular development. The expression levels of the upstream regulator of FoxO3a, Akt, were also examined and it was demonstrated that Akt phosphorylation was significantly reduced in the HFD group compared with the NC group, indicating that Akt/FoxO3a may be also involved in follicular development. Together, the experiments demonstrated that HFD‑induced obesity affected the activity of the Akt/FoxO3a/Smad1/5/8 signaling pathway in a time‑dependent manner during the follicular development of the mouse ovary, leading to abnormal follicular development. These findings may provide part of a theoretical basis for the clinical prevention and treatment of obesity-associated female infertility.
Collapse
Affiliation(s)
- Yanqing Wu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Xinghui Liao
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Lingbin Qi
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Yiping Liu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| |
Collapse
|
49
|
Receptor for advanced glycation end products (RAGE) knockout reduces fetal dysmorphogenesis in murine diabetic pregnancy. Reprod Toxicol 2016; 62:62-70. [DOI: 10.1016/j.reprotox.2016.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 03/07/2016] [Accepted: 04/18/2016] [Indexed: 01/06/2023]
|
50
|
Zhong J, Xu C, Gabbay-Benziv R, Lin X, Yang P. Superoxide dismutase 2 overexpression alleviates maternal diabetes-induced neural tube defects, restores mitochondrial function and suppresses cellular stress in diabetic embryopathy. Free Radic Biol Med 2016; 96:234-44. [PMID: 27130031 PMCID: PMC4912469 DOI: 10.1016/j.freeradbiomed.2016.04.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/21/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Pregestational diabetes disrupts neurulation leading to neural tube defects (NTDs). Oxidative stress resulting from reactive oxygen species (ROS) plays a central role in the induction of NTD formation in diabetic pregnancies. We aimed to determine whether mitochondrial dysfunction increases ROS production leading to oxidative stress and diabetic embryopathy. Overexpression of the mitochondrion-specific antioxidant enzyme superoxide dismutase 2 (SOD2) in a transgenic (Tg) mouse model significantly reduced maternal diabetes-induced NTDs. SOD2 overexpression abrogated maternal diabetes-induced mitochondrial dysfunction by inhibiting mitochondrial translocation of the pro-apoptotic Bcl-2 family members, reducing the number of defective mitochondria in neuroepithelial cells, and decreasing mitochondrial membrane potential. Furthermore, SOD2 overexpression blocked maternal diabetes-increased ROS production by diminishing dihydroethidium staining signals in the developing neuroepithelium, and reducing the levels of nitrotyrosine-modified proteins and lipid hydroperoxide level in neurulation stage embryos. SOD2 overexpression also abolished maternal diabetes-induced endoplasmic reticulum stress. Finally, caspase-dependent neuroepithelial cell apoptosis enhanced by oxidative stress was significantly reduced by SOD2 overexpression. Thus, our findings support the hypothesis that mitochondrial dysfunction in the developing neuroepithelium enhances ROS production, which leads to oxidative stress and endoplasmic reticulum (ER) stress. SOD2 overexpression blocks maternal diabetes-induced oxidative stress and ER stress, and reduces the incidence of NTDs in embryos exposed to maternal diabetes.
Collapse
Affiliation(s)
- Jianxiang Zhong
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Cheng Xu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Rinat Gabbay-Benziv
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Xue Lin
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, United States.
| |
Collapse
|