1
|
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
|
2
|
Chen H, Luo S, Deng X, Li S, Mao Y, Yan J, Cheng Y, Liu X, Pan J, Huang H. Pre-eclamptic foetal programming predisposes offspring to hepatic steatosis via DNA methylation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167189. [PMID: 38648899 DOI: 10.1016/j.bbadis.2024.167189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/01/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVES Gamete and embryo-foetal origins of adult diseases hypothesis proposes that adulthood chronic disorders are associated with adverse foetal and early life traits. Our study aimed to characterise developmental changes and underlying mechanisms of metabolic disorders in offspring of pre-eclampsia (PE) programmed pregnancy. METHODS Nω-Nitro-l-arginine methyl ester hydrochloride (L-NAME) induced pre-eclampsia-like C57BL/6J mouse model was used. Lipid profiling, histological morphology, indirect calorimetry, mRNA sequencing, and pyrosequencing were performed on PE offspring of both young and elderly ages. RESULTS PE offspring exhibited increased postnatal weight gain, hepatic lipid accumulation, enlarged adipocytes, and impaired energy balance that continued to adulthood. Integrated RNA sequencing of foetal and 52-week-old livers revealed that the differentially expressed genes were mainly enriched in lipid metabolism, including glycerol-3-phosphate acyl-transferase 3 (Gpat3), a key enzyme for de novo synthesis of triglycerides (TG), and carnitine palmitoyltransferase-1a (Cpt1a), a key transmembrane enzyme that mediates fatty acid degradation. Pyrosequencing of livers from PE offspring identified hypomethylated and hypermethylated regions in Gpat3 and Cpt1a promoters, which were associated with upregulated and downregulated expressions of Gpat3 and Cpt1a, respectively. These epigenetic alterations are persistent and consistent from the foetal stage to adulthood in PE offspring. CONCLUSION These findings suggest a methylation-mediated epigenetic mechanism for PE-induced intergenerational lipid accumulation, impaired energy balance and obesity in offspring, and indicate the potential benefits of early interventions in offspring exposed to maternal PE to reduce their susceptibility to metabolic disorder in their later life.
Collapse
Affiliation(s)
- Huixi Chen
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; State Key Laboratory of Cardiology, Shanghai 200000, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Sisi Luo
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Xiuyu Deng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Sisi Li
- Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Reproductive Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Jing Yan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Xia Liu
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China.
| | - Hefeng Huang
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Reproductive Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; State Key Laboratory of Cardiology, Shanghai 200000, China.
| |
Collapse
|
3
|
Liu K, Chen Z, Hu W, He B, Xu D, Guo Y, Wang H. Intrauterine developmental origin, programming mechanism, and prevention strategy of fetal-originated hypercholesterolemia. Obes Rev 2024; 25:e13672. [PMID: 38069529 DOI: 10.1111/obr.13672] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/30/2023] [Accepted: 11/05/2023] [Indexed: 02/28/2024]
Abstract
There is increasing evidence that hypercholesterolemia has an intrauterine developmental origin. However, the pathogenesis of fetal-originated is still lacking in a theoretical system, which makes its clinical early prevention and treatment difficult. It has been found that an adverse environment during pregnancy (e.g., xenobiotic exposure) may lead to changes in fetal blood cholesterol levels through changing maternal cholesterol metabolic function and/or placental cholesterol transport function and may also directly affect the liver cholesterol metabolic function of the offspring in utero and continue after birth. Adverse environmental conditions during pregnancy may also raise maternal glucocorticoid levels and promote the placental glucocorticoid barrier opening, leading to fetal overexposure to maternal glucocorticoids. Intrauterine high-glucocorticoid exposure can alter the liver cholesterol metabolism of offspring, resulting in an increased susceptibility to hypercholesterolemia after birth. Abnormal epigenetic modifications are involved in the intrauterine programming mechanism of fetal-originated hypercholesterolemia. Some interventions targeted at pregnant mothers or offspring in early life have been proposed to effectively prevent and treat the development of fetal-originated hypercholesterolemia. In this paper, the recent research progress on fetal-originated hypercholesterolemia was reviewed, with emphasis on intrauterine maternal glucocorticoid programming mechanisms, in order to provide a theoretical basis for its early clinical warning, prevention, and treatment.
Collapse
Affiliation(s)
- Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ze Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bo He
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Dan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
4
|
Zhang Z, Duan Y, Huo J. Lipid Metabolism, Methylation Aberrant, and Osteoporosis: A Multi-omics Study Based on Mendelian Randomization. Calcif Tissue Int 2024; 114:147-156. [PMID: 38071623 DOI: 10.1007/s00223-023-01160-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/03/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Observational studies have shown a causal association between dyslipidemia and osteoporosis, but the genetic causation and complete mechanism of which are uncertain. The disadvantage of previous observational studies is that they are susceptible to confounding factors and bias, that makes it difficult to infer a causal link between those two diseases. Abnormal epigenetic modifications, represented by DNA methylation, are important causes of many diseases. However, there are no studies showing a bridging role for methylation modifications in blood lipid metabolism and osteoporosis. METHODS SNPs for lipid profile (Blood VLDL cholesterol (VLDL-C), blood LDL cholesterol (LDL-C), blood HDL cholesterol (HDL-C), blood triglycerides (TG), diagnosed pure hypercholesterolaemia, blood apolipoprotein B (Apo B), blood apolipoprotein A1(Apo A1)), and bone mineral density (BMD) in different body parts (Heel BMD, lumbar BMD, whole-body BMD, femoral neck BMD) were obtained from large meta-analyses of genome-wide association studies as instrumental variables for two-sample Mendelian randomization. Assessment of the genetic effects of lipid profile-associated methylation sites and bone mineral density was carried out using the summary-data-based Mendelian randomization (SMR) method. RESULTS Two-sample Mendelian randomization showed that there was a negative causal association between hypercholesterolaemia and heel BMD (p = 0.0103, OR = 0.4590), and total body BMD (p = 0.0002, OR = 0.2826). LDL-C had a negative causal association with heel BMD (p = 8.68E-05, OR = 0.9586). VLDL-C had a negative causal association with heel BMD (p = 0.035, OR = 0.9484), lumbar BMD (p = 0.0316, OR = 0.9356), and total body BMD (p = 0.0035, OR = 0.9484). HDL-C had a negative causal association with heel BMD (p = 1.25E-05, OR = 0.9548), lumbar BMD (p = 0.0129, OR = 0.9358), and total body BMD (p = 0.0399, OR = 0.9644). Apo B had a negative causal association with heel BMD (p = 0.0001, OR = 0.9647). Apo A1 had a negative causal association with heel BMD (p = 0.0132, OR = 0.9746) and lumbar BMD (p = 0.0058, OR = 0.9261). The p-values of all positive results corrected by the FDR method remained significant and sensitivity analysis showed that there was no horizontal pleiotropy in the results despite the heterogeneity in some results. SMR identified 3 methylation sites associated with lipid profiles in the presence of genetic effects on BMD: cg15707428(GREB1), cg16000331(SREBF2), cg14364472(NOTCH1). CONCLUSION Our study provides insights into the potential causal links and co-pathogenesis between dyslipidemia and osteoporosis. The genetic effects of dyslipidaemia on osteoporosis may be related to certain aberrant methylation genetic modifications.
Collapse
Affiliation(s)
- ZhaoLiang Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - YuChen Duan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - JianZhong Huo
- Taiyuan Central Hospital, Ninth Hospital of Shanxi Medical University, Southern Fendong Road 256, Taiyuan, 030009, Shanxi, China.
| |
Collapse
|
5
|
Hong J, Tong H, Wang X, Lv X, He L, Yang X, Wang Y, Xu K, Liang Q, Feng Q, Niu T, Niu X, Lu Y. Embryonic diapause due to high glucose is related to changes in glycolysis and oxidative phosphorylation, as well as abnormalities in the TCA cycle and amino acid metabolism. Front Endocrinol (Lausanne) 2023; 14:1135837. [PMID: 38170036 PMCID: PMC10759208 DOI: 10.3389/fendo.2023.1135837] [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: 01/01/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction The adverse effects of high glucose on embryos can be traced to the preimplantation stage. This study aimed to observe the effect of high glucose on early-stage embryos. Methods and results Seven-week-old ICR female mice were superovulated and mated, and the zygotes were collected. The zygotes were randomly cultured in 5 different glucose concentrations (control, 20mM, 40mM, 60mM and 80mM glucose). The cleavage rate, blastocyst rate and total cell number of blastocyst were used to assess the embryo quality. 40 mM glucose was selected to model high glucose levels in this study. 40mM glucose arrested early embryonic development, and the blastocyst rate and total cell number of the blastocyst decreased significantly as glucose concentration was increased. The reduction in the total cell number of blastocysts in the high glucose group was attributed to decreased proliferation and increased cell apoptosis, which is associated with the diminished expression of GLUTs (GLUT1, GLUT2, GLUT3). Furthermore, the metabolic characterization of blastocyst culture was observed in the high-glucose environment. Discussion The balance of glycolysis and oxidative phosphorylation at the blastocyst stage was disrupted. And embryo development arrest due to high glucose is associated with changes in glycolysis and oxidative phosphorylation, as well as abnormalities in the TCA cycle and amino acid metabolism.
Collapse
Affiliation(s)
- Jiewei Hong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongxuan Tong
- Institute of Basic Theory of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuan Wang
- Party Committee Office, Shanxi Health Vocational College, Shanxi, China
| | - Xiaoyan Lv
- Library Collection and Editing Department, Beijing University of Chinese Medicine, Beijing, China
| | - Lijuan He
- Rehabilitation Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xuezhi Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yingli Wang
- Experimental Management Center, Shanxi University of Traditional Chinese Medicine, Shanxi, China
| | - Kaixia Xu
- School of Basic Medicine, Shanxi University of Traditional Chinese Medicine, Shanxi, China
| | - Qi Liang
- Centre for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Qianjin Feng
- Experimental Management Center, Shanxi University of Traditional Chinese Medicine, Shanxi, China
| | - Tingli Niu
- Medical Insurance Office, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xin Niu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Lu
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Sahu DK, Abraham J. Plasma mitochondrial DNA is elevated in maternal serum at first trimester and may serve as a biomarker for prediction of gestational diabetes mellitus. J Diabetes 2023; 15:1095-1102. [PMID: 37658630 PMCID: PMC10755614 DOI: 10.1111/1753-0407.13462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND We evaluated whether an abundance of first-trimester plasma mitochondrial DNA (mtDNA) fragments could predict the risk for the development of gestational diabetes mellitus (GDM) by the late second or early third trimester. METHODS It was a prospective study wherein we enrolled 150 women in their first trimester of gestation. Oral glucose tolerance test (OGTT) was administered both in the first and second trimesters to diagnose GDM. RESULTS Among our cohort, 23 women were diagnosed with GDM in the first trimester and excluded from the study. Of the remaining 127, 29 women were diagnosed with GDM in the second trimester, and 98 women who did not develop GDM served as controls. We amplified blood drawn from each participant during the first trimester for three distinct mtDNA gene sequences: COX, ND4, and D-loop. An abundance of each mtDNA sequence, estimated by the ΔCt method between mtDNA and 18S rRNA, correlated with GDM occurrence in the late second or early third trimester. There was a significant difference in ΔCt COX between controls and those with GDM occurrence in the second trimester (p = .006). These levels were not associated with age or fasting plasma glucose levels in the first trimester. ΔCt COX could predict GDM with a sensitivity of 90% and a specificity of 40%. Though ΔCt ND4 was higher in the GDM-positive group, the levels did not reach statistical significance. ΔCt D-loop was similar in GDM-positive cases and controls who did not develop GDM during pregnancy. CONCLUSIONS These results were in plasma samples collected 3 to 4 months before overt hyperglycemia diagnosis suggestive of GDM. The abundance of plasma mtDNA fragments represents a promising cost-effective, convenient early-stage biomarker for predicting GDM development. Importantly, it can be administered irrespective of the fasting status of the subject. Further assessment of the predictive capacity of these biomarkers within large, diverse populations is needed for effective clinical utility.
Collapse
|
7
|
Chen S, Wang X, Lee BK, Gardner RM. Associations between maternal metabolic conditions and neurodevelopmental conditions in offspring: the mediating effects of obstetric and neonatal complications. BMC Med 2023; 21:422. [PMID: 37936224 PMCID: PMC10631144 DOI: 10.1186/s12916-023-03116-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Maternal pre-gestational diabetes (PGDM), gestational diabetes mellitus (GDM), and overweight/obesity have been associated with increased risks of offspring neurodevelopmental conditions (NDCs) including autism, intellectual disability (ID), and attention deficit/hyperactivity disorder (ADHD). Less is known about whether and how obstetric and neonatal complications (e.g., preterm birth, neonatal asphyxia) could mediate these associations. METHODS In this Swedish register-based cohort study, we examined complications during pregnancy, delivery, and the neonatal period as potential mediators of the relationships between maternal metabolic conditions and offspring NDCs. We quantified the extent to which these obstetric and neonatal factors could mediate the associations of maternal metabolic conditions with offspring NDCs by applying parametric regression models for single mediation analyses and weighting-based methods for multiple mediation analyses under counterfactual frameworks. RESULTS The study sample included 2,352,969 singleton children born to 1,299,692 mothers from 1987-2010 who were followed up until December 31, 2016, of whom 135,832 children (5.8%) were diagnosed with at least one NDC. A substantial portion of the association between maternal PGDM and children's odds of NDCs could be explained by the combined group of obstetric and neonatal complications in the multiple mediation analysis. For instance, these complications explained 44.4% of the relationship between maternal PGDM and offspring ID risk. The proportion of the relationship between maternal overweight/obesity and children's risk of NDCs that could be explained by obstetric and neonatal complications was considerably smaller, ranging from 1.5 to 8.1%. Some complications considered on their own, including pregnancy hypertensive diseases, preterm birth, neonatal asphyxia, and hematological comorbidities, could explain at least 10% of the associations between maternal PGDM and offspring NDCs. Complications during the neonatal period showed a stronger joint mediating effect for the relationship between PGDM and offspring NDCs than those during pregnancy or delivery. CONCLUSIONS Obstetric and neonatal complications could explain nearly half of the association between maternal PGDM and offspring risk of NDCs. The mediating effects were more pronounced for complications during the neonatal period and for specific complications such as pregnancy hypertensive diseases, preterm birth, neonatal asphyxia, and hematological comorbidities. Effective preventive strategies for offspring NDCs should holistically address both the primary metabolic issues related to PGDM and the wide array of potential complications, especially those in the neonatal period.
Collapse
Affiliation(s)
- Shuyun Chen
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.
| | - Xi Wang
- PolicyLab, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brian K Lee
- Department of Epidemiology and Biostatistics, Drexel University School of Public Health, Philadelphia, PA, USA
- A.J. Drexel Autism Institute, Philadelphia, PA, USA
| | - Renee M Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
8
|
Vargas-Soria M, García-Alloza M, Corraliza-Gómez M. Effects of diabetes on microglial physiology: a systematic review of in vitro, preclinical and clinical studies. J Neuroinflammation 2023; 20:57. [PMID: 36869375 PMCID: PMC9983227 DOI: 10.1186/s12974-023-02740-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Diabetes mellitus is a heterogeneous chronic metabolic disorder characterized by the presence of hyperglycemia, commonly preceded by a prediabetic state. The excess of blood glucose can damage multiple organs, including the brain. In fact, cognitive decline and dementia are increasingly being recognized as important comorbidities of diabetes. Despite the largely consistent link between diabetes and dementia, the underlying causes of neurodegeneration in diabetic patients remain to be elucidated. A common factor for almost all neurological disorders is neuroinflammation, a complex inflammatory process in the central nervous system for the most part orchestrated by microglial cells, the main representatives of the immune system in the brain. In this context, our research question aimed to understand how diabetes affects brain and/or retinal microglia physiology. We conducted a systematic search in PubMed and Web of Science to identify research items addressing the effects of diabetes on microglial phenotypic modulation, including critical neuroinflammatory mediators and their pathways. The literature search yielded 1327 records, including 18 patents. Based on the title and abstracts, 830 papers were screened from which 250 primary research papers met the eligibility criteria (original research articles with patients or with a strict diabetes model without comorbidities, that included direct data about microglia in the brain or retina), and 17 additional research papers were included through forward and backward citations, resulting in a total of 267 primary research articles included in the scoping systematic review. We reviewed all primary publications investigating the effects of diabetes and/or its main pathophysiological traits on microglia, including in vitro studies, preclinical models of diabetes and clinical studies on diabetic patients. Although a strict classification of microglia remains elusive given their capacity to adapt to the environment and their morphological, ultrastructural and molecular dynamism, diabetes modulates microglial phenotypic states, triggering specific responses that include upregulation of activity markers (such as Iba1, CD11b, CD68, MHC-II and F4/80), morphological shift to amoeboid shape, secretion of a wide variety of cytokines and chemokines, metabolic reprogramming and generalized increase of oxidative stress. Pathways commonly activated by diabetes-related conditions include NF-κB, NLRP3 inflammasome, fractalkine/CX3CR1, MAPKs, AGEs/RAGE and Akt/mTOR. Altogether, the detailed portrait of complex interactions between diabetes and microglia physiology presented here can be regarded as an important starting point for future research focused on the microglia-metabolism interface.
Collapse
Affiliation(s)
- María Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Mónica García-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Miriam Corraliza-Gómez
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain. .,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
9
|
Tang X, Qin Q, Xu W, Zhang X. Long Non-Coding RNA TUG1 Attenuates Insulin Resistance in Mice with Gestational Diabetes Mellitus via Regulation of the MicroRNA-328-3p/SREBP-2/ERK Axis. Diabetes Metab J 2023; 47:267-286. [PMID: 36653891 PMCID: PMC10040623 DOI: 10.4093/dmj.2021.0216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/09/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have been illustrated to contribute to the development of gestational diabetes mellitus (GDM). In the present study, we aimed to elucidate how lncRNA taurine upregulated gene 1 (TUG1) influences insulin resistance (IR) in a high-fat diet (HFD)-induced mouse model of GDM. METHODS We initially developed a mouse model of HFD-induced GDM, from which islet tissues were collected for RNA and protein extraction. Interactions among lncRNA TUG1/microRNA (miR)-328-3p/sterol regulatory element binding protein 2 (SREBP-2) were assessed by dual-luciferase reporter assay. Fasting blood glucose (FBG), fasting insulin (FINS), homeostasis model assessment of insulin resistance (HOMA-IR), HOMA pancreatic β-cell function (HOMA-β), insulin sensitivity index for oral glucose tolerance tests (ISOGTT) and insulinogenic index (IGI) levels in mouse serum were measured through conducting gain- and loss-of-function experiments. RESULTS Abundant expression of miR-328 and deficient expression of lncRNA TUG1 and SREBP-2 were characterized in the islet tissues of mice with HFD-induced GDM. LncRNA TUG1 competitively bound to miR-328-3p, which specifically targeted SREBP-2. Either depletion of miR-328-3p or restoration of lncRNA TUG1 and SREBP-2 reduced the FBG, FINS, HOMA-β, and HOMA-IR levels while increasing ISOGTT and IGI levels, promoting the expression of the extracellular signal-regulated kinase (ERK) signaling pathway-related genes, and inhibiting apoptosis of islet cells in GDM mice. Upregulation miR-328-3p reversed the alleviative effects of SREBP-2 and lncRNA TUG1 on IR. CONCLUSION Our study provides evidence that the lncRNA TUG1 may prevent IR following GDM through competitively binding to miR-328-3p and promoting the SREBP-2-mediated ERK signaling pathway inactivation.
Collapse
Affiliation(s)
- Xuwen Tang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center Affiliated to Guangzhou Medical University, Guangzhou, China
| | - Qingxin Qin
- Department of Endocrinology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| | - Wenjing Xu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center Affiliated to Guangzhou Medical University, Guangzhou, China
| | - Xuezhen Zhang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center Affiliated to Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Preeclampsia-induced alterations in brain and liver gene expression and DNA methylation patterns in fetal mice. J Dev Orig Health Dis 2023; 14:146-151. [PMID: 35748176 DOI: 10.1017/s2040174422000344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Exposure to pregnancy complications, including preeclampsia (PE), has lifelong influences on offspring's health. We have previously reported that experimental PE, induced in mice by administration of adenoviral sFlt1 at gestational day 8.5 combined with LPS at day 10.5, results in symmetrical growth restriction in female and asymmetrical growth restriction in male offspring. Here, we characterize the molecular phenotype of the fetal brain and liver with respect to gene transcription and DNA methylation at the end of gestation.In fetal brain and liver, expression and DNA methylation of several key regulatory genes is altered by PE exposure, mostly independent of fetal sex. These alterations point toward a decreased gluconeogenesis in the liver and stimulated neurogenesis in the brain, potentially affecting long-term brain and liver function. The observed sex-specific growth restriction pattern is not reflected in the molecular data, showing that PE, rather than tissue growth, drives the molecular phenotype of PE-exposed offspring.
Collapse
|
11
|
Deckmann I, Santos-Terra J, Martel F, Vieira Carletti J. Common pregnancy complications and polyphenols intake: an overview. Crit Rev Food Sci Nutr 2023; 64:5924-5957. [PMID: 36597650 DOI: 10.1080/10408398.2022.2160960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
During pregnancy, the body undergoes a great amount of changes in order to support a healthy developing fetus. In this context, maternal dietary supplementation is widely encouraged to provide adequate nutrition for the newborn. In the past few years, studies have emerged highlighting the benefits of polyphenols intake during pregnancy. Indeed, despite differences among reports, such as experimental model, polyphenol employed, dosage and regimen of administration, there is no doubt that the ingestion of these molecules has a protective effect in relation to three pregnancy-associated diseases or conditions: preeclampsia, gestational diabetes and fetal growth restriction. In this review, we describe the effects of different polyphenols and polyphenol-rich extracts or juices on the main outcomes of these common pregnancy-associated complications, obtained in human, animal and in vitro studies. Therefore, this work provides a critical analysis of the literature, and a summary of evidences, from which future research using polyphenols can be designed and evaluated.
Collapse
Affiliation(s)
- Iohanna Deckmann
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Júlio Santos-Terra
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Jaqueline Vieira Carletti
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
12
|
Bianchi ME, Restrepo JM. Low Birthweight as a Risk Factor for Non-communicable Diseases in Adults. Front Med (Lausanne) 2022; 8:793990. [PMID: 35071274 PMCID: PMC8770864 DOI: 10.3389/fmed.2021.793990] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/08/2021] [Indexed: 11/18/2022] Open
Abstract
According to studies undertaken over the past 40 years, low birthweight (LBW) is not only a significant predictor of perinatal death and morbidity, but also increases the risk of chronic non-communicable diseases (NCDs) in adulthood. The purpose of this paper is to summarize the research on LBW as a risk factor for NCDs in adults. The Barker hypothesis was based on the finding that adults with an LBW or an unhealthy intrauterine environment, as well as a rapid catch-up, die due to NCDs. Over the last few decades, terminology such as thrifty genes, fetal programming, developmental origins of health and disease (DOHaD), and epigenetic factors have been coined. The most common NCDs include cardiovascular disease, diabetes mellitus type 2 (DMT2), hypertension (HT), dyslipidemia, proteinuria, and chronic kidney disease (CKD). Studies in mothers who experienced famine and those that solely reported birth weight as a risk factor for mortality support the concept. Although the etiology of NCD is unknown, Barry Brenner explained the notion of a low glomerular number (nGlom) in LBW children, followed by the progression to hyperfiltration as the physiopathologic etiology of HT and CKD in adults based on Guyton's renal physiology work. Autopsies of several ethnic groups have revealed anatomopathologic evidence in fetuses and adult kidneys. Because of the renal reserve, demonstrating renal function in proportion to renal volume in vivo is more difficult in adults. The greatest impact of these theories can be seen in pediatrics and obstetrics practice.
Collapse
Affiliation(s)
- Maria Eugenia Bianchi
- Laboratory Physiology, Department Basic Sciences, Institute School of Medicine, National Northeast University, Corrientes, Argentina
| | - Jaime M Restrepo
- Department of Pediatrics, Pediatric Nephrology Service, Icesi University, Valle del Lili, Cali, Colombia.,Fundación Valle del Lili, Cali, Colombia
| |
Collapse
|
13
|
Gantenbein KV, Kanaka-Gantenbein C. Highlighting the trajectory from intrauterine growth restriction to future obesity. Front Endocrinol (Lausanne) 2022; 13:1041718. [PMID: 36440208 PMCID: PMC9691665 DOI: 10.3389/fendo.2022.1041718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
During the last decades several lines of evidence reported the association of an adverse intrauterine environment, leading to intrauterine restriction, with future disease, such as obesity and metabolic syndrome, both leading to increased cardiovascular and cancer risk. The underlying explanation for this association has firstly been expressed by the Barker's hypothesis, the "thrifty phenotype hypothesis". According to this hypothesis, a fetus facing an adverse intrauterine environment adapts to this environment through a reprogramming of its endocrine-metabolic status, during the crucial window of developmental plasticity to save energy for survival, providing less energy and nutrients to the organs that are not essential for survival. This theory evolved to the concept of the developmental origin of health and disease (DOHaD). Thus, in the setting of an adverse, f. ex. protein restricted intrauterine environment, while the energy is mainly directed to the brain, the peripheral organs, f.ex. the muscles and the liver undergo an adaptation that is expressed through insulin resistance. The adaptation at the hepatic level predisposes to future dyslipidemia, the modifications at the vascular level to endothelial damage and future hypertension and, overall, through the insulin resistance to the development of metabolic syndrome. All these adaptations are suggested to take place through epigenetic modifications of the expression of genes without change of their amino-acid sequence. The epigenetic modifications leading to future obesity and cardiovascular risk are thought to induce appetite dysregulation, promoting food intake and adipogenesis, facilitating obesity development. The epigenetic modifications may even persist into the next generation even though the subsequent generation has not been exposed to an adverse intrauterine environment, a notion defined as the "transgenerational transfer of environmental information". As a consequence, if the increased public health burden and costs of non-communicable chronic diseases such as obesity, hypertension, metabolic syndrome and type 2 diabetes have to be minimized, special attention should be laid to the healthy lifestyle habits of women of reproductive age, including healthy diet and physical activity to be established long before any pregnancy takes place in order to provide the best conditions for both somatic and mental health of future generations.
Collapse
Affiliation(s)
| | - Christina Kanaka-Gantenbein
- Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, Athens, Greece
- *Correspondence: Christina Kanaka-Gantenbein, ,
| |
Collapse
|
14
|
Schütte T, Kedziora SM, Haase N, Herse F, Alenina N, Müller DN, Bader M, Schupp M, Dechend R, Golic M, Kräker K. Diabetic pregnancy as a novel risk factor for cardiac dysfunction in the offspring-the heart as a target for fetal programming in rats. Diabetologia 2021; 64:2829-2842. [PMID: 34537857 PMCID: PMC8563640 DOI: 10.1007/s00125-021-05566-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/28/2021] [Indexed: 11/02/2022]
Abstract
AIMS/HYPOTHESIS The impact of diabetic pregnancy has been investigated extensively regarding offspring metabolism; however, little is known about the influence on the heart. We aimed to characterise the effects of a diabetic pregnancy on male adult offspring cardiac health after feeding a high-fat diet in an established transgenic rat model. METHODS We applied our rat model for maternal type 2 diabetes characterised by maternal insulin resistance with hyperglycaemia and hyperinsulinaemia. Diabetes was induced preconceptionally via doxycycline-induced knock down of the insulin receptor in transgenic rats. Male wild-type offspring of diabetic and normoglycaemic pregnancies were raised by foster mothers, followed up into adulthood and subgroups were challenged by a high-fat diet. Cardiac phenotype was assessed by innovative speckle tracking echocardiography, circulating factors, immunohistochemistry and gene expression in the heart. RESULTS When feeding normal chow, we did not observe differences in cardiac function, gene expression and plasma brain natriuretic peptide between adult diabetic or normoglycaemic offspring. Interestingly, when being fed a high-fat diet, adult offspring of diabetic pregnancy demonstrated decreased global longitudinal (-14.82 ± 0.59 vs -16.60 ± 0.48%) and circumferential strain (-23.40 ± 0.57 vs -26.74 ± 0.34%), increased relative wall thickness (0.53 ± 0.06 vs 0.37 ± 0.02), altered cardiac gene expression, enlarged cardiomyocytes (106.60 ± 4.14 vs 87.94 ± 1.67 μm), an accumulation of immune cells in the heart (10.27 ± 0.30 vs 6.48 ± 0.48 per fov) and higher plasma brain natriuretic peptide levels (0.50 ± 0.12 vs 0.12 ± 0.03 ng/ml) compared with normoglycaemic offspring on a high-fat diet. Blood pressure, urinary albumin, blood glucose and body weight were unaltered between groups on a high-fat diet. CONCLUSIONS/INTERPRETATION Diabetic pregnancy in rats induces cardiac dysfunction, left ventricular hypertrophy and altered proinflammatory status in adult offspring only after a high-fat diet. A diabetic pregnancy itself was not sufficient to impair myocardial function and gene expression in male offspring later in life. This suggests that a postnatal high-fat diet is important for the development of cardiac dysfunction in rat offspring after diabetic pregnancy. Our data provide evidence that a diabetic pregnancy is a novel cardiac risk factor that becomes relevant when other challenges, such as a high-fat diet, are present.
Collapse
Affiliation(s)
- Till Schütte
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sarah M Kedziora
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nadine Haase
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Herse
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Natalia Alenina
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Dominik N Müller
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Bader
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Michael Schupp
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Dechend
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- HELIOS-Klinikum, Department of Cardiology and Nephrology, Berlin, Germany
| | - Michaela Golic
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- HSD Hochschule Döpfer, University of Applied Sciences, Cologne, Germany
| | - Kristin Kräker
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Experimental and Clinical Research Center - a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
15
|
Intrauterine hyperglycemia impairs memory across two generations. Transl Psychiatry 2021; 11:434. [PMID: 34417446 PMCID: PMC8379206 DOI: 10.1038/s41398-021-01565-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/27/2021] [Accepted: 08/10/2021] [Indexed: 12/22/2022] Open
Abstract
Studies on humans and animals suggest associations between gestational diabetes mellitus (GDM) with increased susceptibility to develop neurological disorders in offspring. However, the molecular mechanisms underpinning the intergenerational effects remain unclear. Using a mouse model of diabetes during pregnancy, we found that intrauterine hyperglycemia exposure resulted in memory impairment in both the first filial (F1) males and the second filial (F2) males from the F1 male offspring. Transcriptome profiling of F1 and F2 hippocampi revealed that differentially expressed genes (DEGs) were enriched in neurodevelopment and synaptic plasticity. The reduced representation bisulfite sequencing (RRBS) of sperm in F1 adult males showed that the intrauterine hyperglycemia exposure caused altered methylated modification of F1 sperm, which is a potential epigenetic mechanism for the intergenerational neurocognitive effects of GDM.
Collapse
|
16
|
Luo H, Lan C, Fan C, Gong X, Chen C, Yu C, Wang J, Luo X, Hu C, Jose PA, Xu Z, Zeng C. Down-regulation of AMPK/PPARδ signalling promotes endoplasmic reticulum stress-induced endothelial dysfunction in adult rat offspring exposed to maternal diabetes. Cardiovasc Res 2021; 118:2304-2316. [PMID: 34415333 PMCID: PMC9890455 DOI: 10.1093/cvr/cvab280] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 01/29/2020] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
AIMS Exposure to maternal diabetes is associated with increased prevalence of hypertension in the offspring. The mechanisms underlying the prenatal programming of hypertension remain unclear. Because endoplasmic reticulum (ER) stress plays a key role in vascular endothelial dysfunction in hypertension, we investigated whether aberrant ER stress causes endothelial dysfunction and high blood pressure in the offspring of dams with diabetes. METHODS AND RESULTS Pregnant Sprague-Dawley rats were intraperitoneally injected with streptozotocin (35 mg/kg) or citrate buffer at Day 0 of gestation. Compared with control mother offspring (CMO), the diabetic mother offspring (DMO) had higher blood pressure and impaired endothelium-dependent relaxation in mesenteric arteries, accompanied by decreased AMPK phosphorylation and PPARδ expression, increased ER stress markers, and reactive oxygen species (ROS) levels. The inhibition of ER stress reversed these aberrant changes in DMO. Ex vivo treatment of mesenteric arteries with an AMPK agonist (A769662) or a PPARδ agonist (GW1516) improved the impaired EDR in DMO and reversed the tunicamycin-induced ER stress, ROS production, and EDR impairment in mesenteric arteries from CMO. The effects of A769662 were abolished by co-treatment with GSK0660 (PPARδ antagonist), whereas the effects of GW1516 were unaffected by Compound C (AMPK inhibitor). CONCLUSION These results suggest an abnormal foetal programming of vascular endothelial function in offspring of rats with maternal diabetes that is associated with increased ER stress, which can be ascribed to down-regulation of AMPK/PPARδ signalling cascade.
Collapse
Affiliation(s)
| | | | | | - Xue Gong
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Cheng Yu
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China,Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Xiaoli Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Cuimei Hu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, 10 Changjiang Branch Rd, Chongqing 400042, P.R. China,Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine and Pharmacology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA,Department of Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Zaicheng Xu
- Corresponding author. Tel: +86 23 68757801; fax: +86 23 68757801, E-mail: (C.Z.); (Z.X.)
| | - Chunyu Zeng
- Corresponding author. Tel: +86 23 68757801; fax: +86 23 68757801, E-mail: (C.Z.); (Z.X.)
| |
Collapse
|
17
|
Vogtmann R, Heupel J, Herse F, Matin M, Hagmann H, Bendix I, Kräker K, Dechend R, Winterhager E, Kimmig R, Köninger A, Gellhaus A. Circulating Maternal sFLT1 (Soluble fms-Like Tyrosine Kinase-1) Is Sufficient to Impair Spiral Arterial Remodeling in a Preeclampsia Mouse Model. Hypertension 2021; 78:1067-1079. [PMID: 34397280 PMCID: PMC8415521 DOI: 10.1161/hypertensionaha.121.17567] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Supplemental Digital Content is available in the text. One driving factor for developing preeclampsia—a pregnancy disorder, often associated with poor spiral artery (SpA)-remodeling and fetal growth restriction—is the anti-angiogenic sFLT1 (soluble fms-like tyrosine kinase-1), which is found to be highly upregulated in preeclampsia patients. The sFLT1-mediated endothelial dysfunction is a common theory for the manifestation of maternal preeclampsia symptoms. However, the influence of sFLT1 on SpA-remodeling and the link between placental and maternal preeclampsia symptoms is less understood. To dissect the hsFLT1 (human sFLT1) effects on maternal and/or fetoplacental physiology in preeclampsia, sFLT1-transgenic mice with systemic hsFLT1 overexpression from midgestation onwards were used. SpA-remodeling was analyzed on histological and molecular level in placental/mesometrial triangle tissues. Maternal kidney and aorta morphology was investigated, combined with blood pressure measurements via telemetry. hsFLT1 overexpression resulted in maternal hypertension, aortic wall thickening, and elastin breakdown. Furthermore, maternal kidneys showed glomerular endotheliosis, podocyte damage, and proteinuria. preeclampsia symptoms were combined with fetal growth restriction already at the end of the second trimester and SpA-remodeling was strongly impaired as shown by persisted vascular smooth muscle cells. This phenotype was associated with shallow trophoblast invasion, delayed presence of uterine natural killer cells, and altered lymphatic angiogenesis. Overall, this study showed that circulating maternal hsFLT1 is sufficient to induce typical maternal preeclampsia-like symptoms in mice and impair the SpA-remodeling independent from the fetoplacental compartment, revealing new insights into the interaction between the placental and maternal contribution of preeclampsia.
Collapse
Affiliation(s)
- Rebekka Vogtmann
- Department of Gynecology and Obstetrics, University Hospital Essen, Germany (R.V., J.H., R.K., A.K., A.G.)
| | - Jacqueline Heupel
- Department of Gynecology and Obstetrics, University Hospital Essen, Germany (R.V., J.H., R.K., A.K., A.G.)
| | - Florian Herse
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), Berlin, Germany (F.H., K.K., R.D.).,Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany (F.H., K.K.)
| | - Mahsa Matin
- Department II of Internal Medicine-Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine-University Hospital Cologne, Cologne, Germany and Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases, Germany (M.M., H.H.)
| | - Henning Hagmann
- Department II of Internal Medicine-Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine-University Hospital Cologne, Cologne, Germany and Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases, Germany (M.M., H.H.)
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University of Duisburg-Essen, Germany (I.B.)
| | - Kristin Kräker
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), Berlin, Germany (F.H., K.K., R.D.).,Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany (F.H., K.K.).,Charité-Universitätsmedizin Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (K.K.)
| | - Ralf Dechend
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), Berlin, Germany (F.H., K.K., R.D.).,HELIOS Klinikum, Berlin, Germany (R.D.)
| | - Elke Winterhager
- Imaging Center Essen, EM Unit, University Hospital Essen, Germany (E.W.)
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, Germany (R.V., J.H., R.K., A.K., A.G.)
| | - Angela Köninger
- Department of Gynecology and Obstetrics, University Hospital Essen, Germany (R.V., J.H., R.K., A.K., A.G.).,Department of Gynecology and Obstetrics, Clinic of the Order of St. John, St. Hedwigs Clinic, Regensburg, Germany (A.K.)
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University Hospital Essen, Germany (R.V., J.H., R.K., A.K., A.G.)
| |
Collapse
|
18
|
Haase N, Foster DJ, Cunningham MW, Bercher J, Nguyen T, Shulga-Morskaya S, Milstein S, Shaikh S, Rollins J, Golic M, Herse F, Kräker K, Bendix I, Serdar M, Napieczynska H, Heuser A, Gellhaus A, Thiele K, Wallukat G, Müller DN, LaMarca B, Dechend R. RNA interference therapeutics targeting angiotensinogen ameliorate preeclamptic phenotype in rodent models. J Clin Invest 2021; 130:2928-2942. [PMID: 32338644 DOI: 10.1172/jci99417] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/13/2020] [Indexed: 01/03/2023] Open
Affiliation(s)
- Nadine Haase
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin Germany.,Experimental and Clinical Research Center, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | | | - Mark W Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Julia Bercher
- Experimental and Clinical Research Center, Berlin, Germany
| | - Tuyen Nguyen
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | | | | | - Jeff Rollins
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Michaela Golic
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin Germany.,Experimental and Clinical Research Center, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Florian Herse
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin Germany.,Experimental and Clinical Research Center, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Kristin Kräker
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin Germany.,Experimental and Clinical Research Center, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Ivo Bendix
- Department of Pediatrics I Neonatology and Experimental Perinatal Neurosciences and
| | - Meray Serdar
- Department of Pediatrics I Neonatology and Experimental Perinatal Neurosciences and
| | - Hanna Napieczynska
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Arnd Heuser
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kristin Thiele
- Department of Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerd Wallukat
- Experimental and Clinical Research Center, Berlin, Germany
| | - Dominik N Müller
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin Germany.,Experimental and Clinical Research Center, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA.,Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ralf Dechend
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin Germany.,Experimental and Clinical Research Center, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,HELIOS-Klinikum, Berlin, Germany.Preeclampsia, with the hallmark features of new-onset hypertension and proteinuria after 20 weeks of gestation, is a major cause of fetal and maternal morbidity and mortality. Studies have demonstrated a role for the renin-angiotensin system (RAS) in its pathogenesis; however, small-molecule RAS blockers are contraindicated because of fetal toxicity. We evaluated whether siRNA targeting maternal hepatic angiotensinogen (Agt, ) could ameliorate symptoms of preeclampsia without adverse placental or fetal effects in 2 rodent models. The first model used a cross of females expressing human Agt, with males expressing human renin, resulting in upregulation of the circulating and uteroplacental RAS. The second model induced ischemia/reperfusion injury and subsequent local and systemic inflammation by surgically reducing placental blood flow mid-gestation (reduced uterine perfusion pressure [RUPP]). These models featured hypertension, proteinuria, and fetal growth restriction, with altered biomarkers. siRNA treatment ameliorated the preeclamptic phenotype in both models, reduced blood pressure, and improved intrauterine growth restriction, with no observed deleterious effects on the fetus. Treatment also improved the angiogenic balance and proteinuria in the transgenic model, and it reduced angiotensin receptor activating antibodies in both. Thus, an RNAi therapeutic targeting Agt, ameliorated the clinical sequelae and improved fetal outcomes in 2 rodent models of preeclampsia
| |
Collapse
|
19
|
Lurbe E, Ingelfinger J. Developmental and Early Life Origins of Cardiometabolic Risk Factors: Novel Findings and Implications. Hypertension 2021; 77:308-318. [PMID: 33390043 DOI: 10.1161/hypertensionaha.120.14592] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The intent of this review is to critically consider the data that support the concept of programming and its implications. Birth weight and growth trajectories during childhood are associated with cardiometabolic disease in adult life. Both extremes, low and high birth weight coupled with postnatal growth increase the early presence of cardiometabolic risk factors and vascular imprinting, crucial elements of this framework. Data coming from epigenetics, proteomics, metabolomics, and microbiota added relevant information and contribute to better understanding of mechanisms as well as development of biomarkers helping to move forward to take actions. Research has reached a stage in which sufficiently robust data calls for new initiatives focused on early life. Prevention starting early in life is likely to have a very large impact on reducing disease incidence and its associated effects at the personal, economic, and social levels.
Collapse
Affiliation(s)
- Empar Lurbe
- From the Pediatric Department, Consorcio Hospital General, University of Valencia (E.L.)
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, Spain (E.L.)
| | - Julie Ingelfinger
- Department of Pediatrics, Harvard Medical School, Mass General Hospital for Children, Massachusetts General Hospital, Boston (J.I.)
| |
Collapse
|
20
|
Schütte T, Kedziora SM, Haase N, Herse F, Busjahn A, Birukov A, Alenina N, Müller DN, Bader M, Schupp M, Dechend R, Kräker K, Golic M. Intrauterine Exposure to Diabetic Milieu Does Not Induce Diabetes and Obesity in Male Adulthood in a Novel Rat Model. Hypertension 2020; 77:202-215. [PMID: 33249866 DOI: 10.1161/hypertensionaha.120.16360] [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] [Indexed: 02/06/2023]
Abstract
Several studies show an association of maternal diabetes during pregnancy with adverse offspring metabolic health. Other studies, however, suggest that this effect might be biased by obesity, which is independently associated with offspring metabolic disease and often coexistent to maternal diabetes. We performed a prospective study in a rat model to test the hypothesis that the burden of a diabetic pregnancy without obesity deteriorates metabolic health in male offspring. We generated maternal type 2 diabetes before conception that persisted during pregnancy by knockdown of the insulin receptor in small hairpin RNA-expressing transgenic rats. Male WT (wild type) offspring were followed up until adulthood and metabolically challenged by high-fat diet. Blood glucose was measured continuously via a telemetry device. Glucose and insulin tolerance tests were performed, and body composition was analyzed. Weight gain and glucose levels during adolescence and adulthood were similar in male offspring of diabetic and control pregnancies. Body weight and fat mass after high-fat diet, as well as glucose and insulin tolerance tests, were unaltered between male adult offspring of both groups. Glycemic control consisting of up to 49 000 individual glucose measures was comparable between both groups. Intrauterine exposure to maternal hyperglycemia and hyperinsulinemia without obesity had no impact on male offspring metabolic health in our model. We conclude that the intrauterine exposure itself does not represent a mechanism for fetal programming of diabetes and obesity in our model. Other maternal metabolic parameters during pregnancy, such as obesity, might impact long-term offspring metabolic health.
Collapse
Affiliation(s)
- Till Schütte
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Institute of Pharmacology, Berlin, Germany (T.S., M.S.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.)
| | - Sarah M Kedziora
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.)
| | - Nadine Haase
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.)
| | - Florian Herse
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.)
| | | | - Anna Birukov
- German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.).,Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany (A. Birukov).,German Center for Diabetes Research, München-Neuherberg, Germany (A. Birukov)
| | - Natalia Alenina
- Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.)
| | - Dominik N Müller
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.)
| | - Michael Bader
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Institute for Biology, University of Lübeck, Germany (M.B.)
| | - Michael Schupp
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Institute of Pharmacology, Berlin, Germany (T.S., M.S.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.)
| | - Ralf Dechend
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.).,HELIOS Klinikum, Department of Cardiology and Nephrology, Berlin, Germany (R.D.)
| | - Kristin Kräker
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.)
| | - Michaela Golic
- From the Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., D.N.M., M.B., M.S., R.D., K.K., M.G.).,Berlin Institute of Health, Germany (T.S., S.M.K., N.H., F.H., N.A., D.N.M., M.B., M.S., R.D., K.K., M.G.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (T.S., S.M.K., N.H., A. Birukov, N.A., D.N.M., M.B., R.D., K.K., M.G.).,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (S.M.K., N.H., F.H., N.A., D.N.M., M.B., R.D., K.K., M.G.).,Experimental and Clinical Research Center-a joint cooperation between the Max-Delbrück-Center for Molecular Medicine and the Charité-Universitätsmedizin Berlin, Germany (S.M.K., N.H., F.H., A. Birukov, D.N.M., R.D., K.K., M.G.)
| |
Collapse
|
21
|
Shub A, Lappas M. Pregestational diabetes in pregnancy: Complications, management, surveillance, and mechanisms of disease-A review. Prenat Diagn 2020; 40:1092-1098. [PMID: 32333803 DOI: 10.1002/pd.5718] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/12/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
Abstract
Diabetes is an increasingly common diagnosis among pregnant women. Pregestational diabetes is associated with an increase in many adverse pregnancy outcomes, which impact both on the woman and her fetus. The models of pregnancy care for women with diabetes are based largely on observational data or consensus opinion. Strategies for aneuploidy screening and monitoring for fetal well-being should be modified in women with diabetes. There is an increasing understanding of the mechanisms by which congenital anomalies and disorders of fetal growth occur, involving epigenetic modifications, changes in gene expression in critical developmental pathways, and oxidative stress. This knowledge may lead to pathways for improved care for these high-risk pregnancies.
Collapse
Affiliation(s)
- Alexis Shub
- Department of Obstetrics and Gynecology, University of Melbourne, Parkville, Australia.,Perinatal Department, Mercy Hospital for Women, Heidelberg, Australia
| | - Martha Lappas
- Department of Obstetrics and Gynecology, University of Melbourne, Parkville, Australia
| |
Collapse
|
22
|
Arima Y, Fukuoka H. Developmental origins of health and disease theory in cardiology. J Cardiol 2020; 76:14-17. [PMID: 32115330 DOI: 10.1016/j.jjcc.2020.02.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 01/29/2020] [Accepted: 02/09/2020] [Indexed: 01/17/2023]
Abstract
Numerous epidemiological and animal studies disclosed that birth weight is inversely associated with the incidence of the lifestyle-related disorders in adult life, such as cardiovascular disease, diabetes, and /or chronic kidney disease. Lower birth weight occurs in numerous undesired intrauterine environments including malnutrition, smoking, alcohol consumption, or stress. The Developmental Origin of Health and Disease (DOHaD) theory is based on the concept that the origins of lifestyle-related disease is formed at the time of fertilization, embryonic, fetal, and neonatal stages by the interrelation between genes and the environments (nutrition, stress, or environmental chemicals). Adult disease develops after delivery facing to abnormal environments such as over-nutrition, much stress, or lack of exercise. Disease develops through these two insults. This concept was first proposed as the "Barker Hypothesis." David Barker had discovered the relation between the lower birth weight and the higher prevalence of ischemic heart disease mortality. Previous epidemiologic studies have found the people exposed to famine during early life had higher risks of cardiovascular diseases in adulthood. Yet, the exact mechanisms that permanently change the structure, physiology, and endocrine status of an individual across their lifespan following altered growth during fetal life are not entirely clear. Epidemiological studies including prospective cohort and observational analysis of the people exposed to malnutrition during fetal or infancy have disclosed the strong relation between the lower birth weight and the higher cardiovascular risks in adults. Recent progress of epigenetic studies unveiled strong genetic association. Hormonal regulation and epigenetic modifications have an important role for proper organ development and physiological functions. The molecular mechanism of predisposition is supposed to be the epigenetics modifications. Their dysregulation is related to the acquisition of the disease-susceptible trait. In this review, we overview the concept of DOHaD and introduce related clinical and basic research.
Collapse
Affiliation(s)
- Yuichiro Arima
- Department of Cardiovascular Medicine, Kumamoto University, Kumamoto City, Japan; International Research Center for Medical Science, Kumamoto University, Kumamoto City, Japan.
| | - Hideoki Fukuoka
- Department of Progressive DOHaD Research, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
23
|
Matthes S, Mosienko V, Popova E, Rivalan M, Bader M, Alenina N. Targeted Manipulation of Brain Serotonin: RNAi-Mediated Knockdown of Tryptophan Hydroxylase 2 in Rats. ACS Chem Neurosci 2019; 10:3207-3217. [PMID: 30977636 DOI: 10.1021/acschemneuro.8b00635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tryptophan hydroxylase (TPH) is the rate-limiting enzyme in the biosynthesis of the biogenic monoamine serotonin (5-hydroxytryptamine, 5-HT). Two existing TPH isoforms are responsible for the generation of two distinct serotonergic systems in vertebrates. TPH1, predominantly expressed in the gastrointestinal tract and pineal gland, mediates 5-HT biosynthesis in non-neuronal tissues, while TPH2, mainly found in the raphe nuclei of the brain stem, is accountable for the production of 5-HT in the brain. Neuronal 5-HT is a key regulator of mood and behavior and its deficiency has been implicated in a variety of neuropsychiatric disorders, e.g., depression and anxiety. To gain further insights into the complexity of central 5-HT modulations of physiological and pathophysiological processes, a new transgenic rat model, allowing an inducible gene knockdown of Tph2, was established based on doxycycline-inducible shRNA-expression. Biochemical phenotyping revealed a functional knockdown of Tph2 mRNA expression following oral doxycycline administration, with subsequent reductions in the corresponding levels of TPH2 enzyme expression and activity. Transgenic rats showed also significantly decreased tissue levels of 5-HT and its degradation product 5-Hydroxyindoleacetic acid (5-HIAA) in the raphe nuclei, hippocampus, hypothalamus, and cortex, while peripheral 5-HT concentrations in the blood remained unchanged. In summary, this novel transgenic rat model allows inducible manipulation of 5-HT biosynthesis specifically in the brain and may help to elucidate the role of 5-HT in the pathophysiology of affective disorders.
Collapse
Affiliation(s)
- Susann Matthes
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Valentina Mosienko
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- College of Medicine and Health, Institute of Biomedical and Clinical Sciences, University of Exeter, Hatherly Building, Prince of Wales Rd., EX4 4PS Exeter, United Kingdom
| | - Elena Popova
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
| | - Marion Rivalan
- Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany
- Institute of Translational Biomedicine, St. Petersburg State University, Saint Petersburg 199034, Russia
| |
Collapse
|
24
|
Falkner B. Too Much Sugar Is Not Good for the Fetus. Am J Hypertens 2019; 32:329-330. [PMID: 30772909 DOI: 10.1093/ajh/hpz025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 11/14/2022] Open
|
25
|
Langmia IM, Kräker K, Weiss SE, Haase N, Schütte T, Herse F, Dechend R. Cardiovascular Programming During and After Diabetic Pregnancy: Role of Placental Dysfunction and IUGR. Front Endocrinol (Lausanne) 2019; 10:215. [PMID: 31024453 PMCID: PMC6466995 DOI: 10.3389/fendo.2019.00215] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/18/2019] [Indexed: 12/31/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a condition whereby a fetus is unable to achieve its genetically determined potential size. IUGR is a global health challenge due to high mortality and morbidity amongst affected neonates. It is a multifactorial condition caused by maternal, fetal, placental, and genetic confounders. Babies born of diabetic pregnancies are usually large for gestational age but under certain conditions whereby prolonged uncontrolled hyperglycemia leads to placental dysfunction, the outcome of the pregnancy is an intrauterine growth restricted fetus with clinical features of malnutrition. Placental dysfunction leads to undernutrition and hypoxia, which triggers gene modification in the developing fetus due to fetal adaptation to adverse utero environmental conditions. Thus, in utero gene modification results in future cardiovascular programming in postnatal and adult life. Ongoing research aims to broaden our understanding of the molecular mechanisms and pathological pathways involved in fetal programming due to IUGR. There is a need for the development of effective preventive and therapeutic strategies for the management of growth-restricted infants. Information on the mechanisms involved with in utero epigenetic modification leading to development of cardiovascular disease in adult life will increase our understanding and allow the identification of susceptible individuals as well as the design of targeted prevention strategies. This article aims to systematically review the latest molecular mechanisms involved in the pathogenesis of IUGR in cardiovascular programming. Animal models of IUGR that used nutrient restriction and hypoxia to mimic the clinical conditions in humans of reduced flow of nutrients and oxygen to the fetus will be discussed in terms of cardiac remodeling and epigenetic programming of cardiovascular disease. Experimental evidence of long-term fetal programming due to IUGR will also be included.
Collapse
Affiliation(s)
- Immaculate M. Langmia
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Alexander von Humboldt Foundation, Bonn, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sara E. Weiss
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Till Schütte
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Center for Cardiovascular Research, Institute of Pharmacology, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- HELIOS-Klinikum, Berlin, Germany
- *Correspondence: Ralf Dechend
| |
Collapse
|
26
|
Adverse neuropsychiatric development following perinatal brain injury: from a preclinical perspective. Pediatr Res 2019; 85:198-215. [PMID: 30367160 DOI: 10.1038/s41390-018-0222-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/11/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023]
Abstract
Perinatal brain injury is a leading cause of death and disability in young children. Recent advances in obstetrics, reproductive medicine and neonatal intensive care have resulted in significantly higher survival rates of preterm or sick born neonates, at the price of increased prevalence of neurological, behavioural and psychiatric problems in later life. Therefore, the current focus of experimental research shifts from immediate injury processes to the consequences for brain function in later life. The aetiology of perinatal brain injury is multi-factorial involving maternal and also labour-associated factors, including not only placental insufficiency and hypoxia-ischaemia but also exposure to high oxygen concentrations, maternal infection yielding excess inflammation, genetic factors and stress as important players, all of them associated with adverse long-term neurological outcome. Several animal models addressing these noxious stimuli have been established in the past to unravel the underlying molecular and cellular mechanisms of altered brain development. In spite of substantial efforts to investigate short-term consequences, preclinical evaluation of the long-term sequelae for the development of cognitive and neuropsychiatric disorders have rarely been addressed. This review will summarise and discuss not only current evidence but also requirements for experimental research providing a causal link between insults to the developing brain and long-lasting neurodevelopmental disorders.
Collapse
|
27
|
Scioscia M. Relevant Fetal Epigenetic Modifications Result From a Diabetic Intrauterine Environment: Healthy Aging Starts With a Healthy Pregnancy. Hypertension 2018; 71:822-823. [PMID: 29610264 DOI: 10.1161/hypertensionaha.118.10868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Marco Scioscia
- From the Department of Obstetrics and Gynecology, Sacro Cuore Don Calabria Hospital, Verona, Italy and Department of Obstetrics and Gynecology, Policlinico Hospital of Abano Terme, Padova, Italy.
| |
Collapse
|
28
|
Golic M, Kräker K, Fischer C, Alenina N, Haase N, Herse F, Schütte T, Henrich W, Müller DN, Busjahn A, Bader M, Dechend R. Continuous Blood Glucose Monitoring Reveals Enormous Circadian Variations in Pregnant Diabetic Rats. Front Endocrinol (Lausanne) 2018; 9:271. [PMID: 29896157 PMCID: PMC5986873 DOI: 10.3389/fendo.2018.00271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/09/2018] [Indexed: 11/13/2022] Open
Abstract
AIM Diabetes in pregnancy is a major burden with acute and long-term consequences. Its treatment requires adequate diagnosis and monitoring of therapy. Many experimental research on diabetes during pregnancy has been performed in rats. Recently, continuous blood glucose monitoring of non-pregnant diabetic rats revealed an increased circadian variability of blood glucose that made a single blood glucose measurement per day inappropriate to reflect glycemic status. Continuous blood glucose measurement has never been performed in pregnant rats. We wanted to perform continuous blood glucose monitoring in pregnant rats to decipher the influence of pregnancy on blood glucose in diabetic and normoglycemic status. METHODS We used the transgenic Tet29 diabetes rat model with an inducible knock down of the insulin receptor via RNA interference upon application of doxycycline (DOX) leading to insulin resistant type II diabetes. All Tet29 rats received a HD-XG telemetry implant (Data Sciences International, USA) that measured blood glucose and activity continuously. Rats were divided into four groups and blood glucose was monitored until end of pregnancy or the corresponding period: Tet29 + DOX (diabetic) non-pregnant, Tet29 + DOX (diabetic) pregnant, Tet29 (normoglycemic) non-pregnant, Tet29 (normoglycemic) pregnant. RESULTS All analyzed rats displayed a circadian variation in blood glucose concentration. Circadian variability was much more pronounced in pregnant diabetic rats than in normoglycemic pregnant rats. Pregnancy ameliorated variation in blood glucose in diabetic situation. Pregnancy continuously decreased blood glucose during normoglycemic pregnancy. Diabetic rats were less active than normoglycemic rats. We performed a calculation showing that application of continuous blood glucose measurement reduces animal numbers needed to detect a given effect in experimental setting by decreasing variability and SD. INTERPRETATION Continuous blood glucose monitoring via a telemetry device in pregnant rats provides a more informative picture of the glycemic situation in comparison to single measurements. This could improve diagnosis and therapy of diabetes, decrease animal numbers within experimental settings, and add another physiological parameter (activity) to the analysis that could be helpful in testing therapeutic concepts targeting blood glucose levels and peripheral muscle function. We propose continuous glucose monitoring as a new tool for the evaluation of pregnant diabetic rats.
Collapse
Affiliation(s)
- Michaela Golic
- Department of Obstetrics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Gynecology With Breast Center, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Partner Site Berlin, Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Berlin, Germany
| | - Caroline Fischer
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Frankfurt, Germany
| | - Natalia Alenina
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Partner Site Berlin, Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Partner Site Berlin, Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Till Schütte
- Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Pharmacology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Wolfgang Henrich
- Department of Obstetrics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dominik N. Müller
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Partner Site Berlin, Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Berlin, Germany
| | | | - Michael Bader
- Berlin Institute of Health (BIH), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Partner Site Berlin, Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Klinikum Berlin, Berlin, Germany
- *Correspondence: Ralf Dechend,
| |
Collapse
|