1
|
Diniz F, Edgington-Giordano F, El-Dahr SS, Tortelote GG. Early metabolic and hemodynamic indicators of kidney dysfunction in mice offspring from parental low protein diet. Mol Cell Pediatr 2024; 11:11. [PMID: 39407060 PMCID: PMC11480283 DOI: 10.1186/s40348-024-00184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Parental malnutrition, particularly a low-protein diet (LPD), causes oligonephropathy at birth and predisposes offspring to hypertension and chronic kidney disease later in life. The onset of adult kidney disease varies based on genetics and environmental factors, often with subclinical alterations in kidney function being overlooked. This study aimed to examine changes in kidney morphology before significant kidney function decline in the offspring of mice fed a low-protein diet. METHODS Using a combination of histological analysis, kidney metabolic and hemodynamic panel assessments, and advanced statistical techniques such as Linear Discriminant Analysis (LDA) and Principal Component Analysis (PCA), we investigated the initial impact of a maternal low-protein diet (LPD) on kidney development and function. Our study utilized 12-week-old F1 mice from F0 parents fed either a low-protein diet (LPD) or a normal-protein diet (NPD) before the onset of hypertension. RESULTS The offspring (F1 generation) of parents (F0 generation) fed an LPD show reduced body weight from birth to P20. The kidney weight was also reduced compared to F1 offspring from parents fed an NPD. At 12 weeks of age, body weight normalized, but kidney weight remained low. Offspring of parents fed an LPD displayed abnormal kidney morphology, including dilated tubules, oligonephropathy, and fluid-filled cysts which had worsened with age. A kidney metabolic panel analysis at 12 weeks revealed a slight but consistent increase in urine albumin, plasma creatinine, mean urea, and BUN concentrations. Although no significant changes in hemodynamic variables were observed, 2/12 mice, both males, showed alterations in systolic blood pressure, suggesting sex-specific effects when comparing F1 mice from F0 fed either diet. Overall, kidney metabolic changes were strongly correlated to parental LPD. CONCLUSION Our findings indicate that significant kidney damage must accumulate in the F1 generation from parents fed an LPD before any detectable changes in blood pressure occur. Our study suggests that small variations in kidney metabolic function may point to early kidney damage and should not be overlooked in the offspring of these malnourished mice and likely humans.
Collapse
Affiliation(s)
- Fabiola Diniz
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Francesca Edgington-Giordano
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Samir S El-Dahr
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Giovane G Tortelote
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
2
|
Andreescu M. Correlation Between Maternal-Fetus Interface and Placenta-Mediated Complications. Cureus 2024; 16:e62457. [PMID: 38882223 PMCID: PMC11180486 DOI: 10.7759/cureus.62457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2024] [Indexed: 06/18/2024] Open
Abstract
Pregnancy is a highly regulated biological phenomenon that involves the development of a semi-allogeneic fetus inside the uterus of the mother. The maternal-fetal interface is a critical junction where communication takes place between the fetal and maternal immune systems, which determine the outcome of the pregnancy. The interface is composed of the decidua and placenta. The main cells present at the maternal-fetal interface include invading trophoblasts, maternal immune cells, and decidual stromal cells. Although maternal tolerance is crucial for maintaining a successful pregnancy, the role of the placenta in pregnancy is also important. Dysregulation of the placenta leads to various placenta-mediated complications, such as preeclampsia, intrauterine growth restriction, and placental abruption. Although the exact mechanism involving these complications is unclear, research has elucidated various factors involved in these pregnancy disorders. This review aimed to provide a summary of the maternal-fetal interface and immune mechanisms involved in placenta-mediated complications.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Faculty of Medicine, Titu Maiorescu University, Bucharest, ROU
- Hematology, Colentina Clinical Hospital, Bucharest, ROU
| |
Collapse
|
3
|
Ao Z, Wu Z, Hu G, Gong T, Zhang C, Yang Z, Zhang Y. Implications for miR-339-5p regulation of trophoblast proliferation and migration in placentas associated with porcine intrauterine growth retardation using integrated transcriptome sequencing analysis. Theriogenology 2024; 216:127-136. [PMID: 38181538 DOI: 10.1016/j.theriogenology.2023.12.035] [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: 09/01/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
Placental dysfunction is considered as one of the main etiologies of fetal intrauterine growth retardation (IUGR). MicroRNAs (miRNAs) have been demonstrated to be a vital epigenetic modification involved in regulating the placental function and pregnancy outcomes in mammals. However, the mechanisms underlying placenta-specific miRNAs involved in the occurrence and development of pig IUGR remain unclear. In this work, we compared the placental morphologies of piglets with IUGR and normal birth weight (NBW) by using histomorphological analysis and performed a miRNA-mRNA integrative analysis of the gene expression profiles of IUGR and NBW placentas through RNA sequencing. We also investigated the role of differentially expressed ssc-miR-339-5p/GRIK3 through an in vitro experiment on porcine trophoblast cells (PTr2). IUGR piglets had significantly lower birth weight, placental weight, placental efficiency, and placental villus and capillary densities compared with the NBW piglets (P < 0.05). A total of 81 differentially expressed miRNAs and 726 differentially expressed genes in the placentas were screened out between the IUGR and NBW groups. The miRNA-mRNA interaction networks revealed the key core miRNA (ssc-miR-339-5p) and its corresponding target genes. Subsequently, we found that upregulation of ssc-miR-339-5p significantly inhibited the migration and proliferation of PTr2 cells (P < 0.05). The dual-luciferase reporter system showed that GRIK3 was the target gene of ssc-miR-339-5p, and the transcription level of GRIK3 may be negatively regulated by ssc-miR-339-5p. Additionally, overexpression of ssc-miR-339-5p significantly increased (P < 0.05) the mRNA expression levels of genes involved in the cytokine-cytokine receptor interaction pathway. These results indicate that ssc-miR-339-5p may affect the migration and proliferation of trophoblast cells by regulating the expression of GRIK3 and altering the placental inflammatory response, resulting in a suboptimal morphology and function of the placenta and the development of pig IUGR.
Collapse
Affiliation(s)
- Zheng Ao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Zhimin Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Guangling Hu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Ting Gong
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Caizai Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Zhenqing Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Yiyu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
4
|
Alsharairi NA, Li L. Gut Microbiota, Inflammation, and Probiotic Supplementation in Fetal Growth Restriction-A Comprehensive Review of Human and Animal Studies. Life (Basel) 2023; 13:2239. [PMID: 38137841 PMCID: PMC10745050 DOI: 10.3390/life13122239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Fetal growth restriction (FGR) is a pathological state that represents a fetus's inability to achieve adequate growth during pregnancy. Several maternal, placental, and fetal factors are likely associated with FGR etiology. FGR is linked to severe fetal and neonatal complications, as well as adverse health consequences in adulthood. Numerous randomized controlled trials (RCTs) have demonstrated improved growth in FGR fetuses with promising treatment strategies such as maternal micronutrient, amino acid, and nitric oxide supplementation. Elevated inflammation in pregnant women diagnosed with FGR has been associated with an imbalance between pro- and anti-inflammatory cytokines. Gut microbiota dysbiosis may result in increased FGR-related inflammation. Probiotic treatment may relieve FGR-induced inflammation and improve fetal growth. The aim of this review is to provide an overview of the gut microbiota and inflammatory profiles associated with FGR and explore the potential of probiotics in treating FGR.
Collapse
Affiliation(s)
- Naser A. Alsharairi
- Heart, Mind and Body Research Group, Griffith University, Gold Coast, QLD 4222, Australia
| | - Li Li
- School of Science, Western Sydney University, Richmond, NSW 2753, Australia;
| |
Collapse
|
5
|
Wang W, Chen S, Qiao L, Zhang S, Liu Q, Yang K, Pan Y, Liu J, Liu W. Four Markers Useful for the Distinction of Intrauterine Growth Restriction in Sheep. Animals (Basel) 2023; 13:3305. [PMID: 37958061 PMCID: PMC10648371 DOI: 10.3390/ani13213305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/14/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Intrauterine growth restriction (IUGR) is a common perinatal complication in animal reproduction, with long-lasting negative effects on neonates and postnatal animals, which seriously negatively affects livestock production. In this study, we aimed to identify potential genes associated with the diagnosis of IUGR through bioinformatics analysis. Based on the 73 differentially expressed related genes obtained by differential analysis and weighted gene co-expression network analysis, we used three machine learning algorithms to identify 4 IUGR-related hub genes (IUGR-HGs), namely, ADAM9, CRYL1, NDP52, and SERPINA7, whose ROC curves showed that they are a good diagnostic target for IUGR. Next, we identified two molecular subtypes of IUGR through consensus clustering analysis and constructed a gene scoring system based on the IUGR-HGs. The results showed that the IUGR score was positively correlated with the risk of IUGR. The AUC value of IUGR scoring accuracy was 0.970. Finally, we constructed a new artificial neural network model based on the four IUGR-HGs to diagnose sheep IUGR, and its accuracy reached 0.956. In conclusion, the IUGR-HGs we identified provide new potential molecular markers and models for the diagnosis of IUGR in sheep; they can better diagnose whether sheep have IUGR. The present findings provide new perspectives on the diagnosis of IUGR.
Collapse
Affiliation(s)
- Wannian Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
| | - Sijia Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
| | - Liying Qiao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
| | - Siying Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
| | - Qiaoxia Liu
- Shanxi Animal Husbandry Technology Extension Service Center, Taiyuan 030001, China;
| | - Kaijie Yang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
| | - Yangyang Pan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
| | - Jianhua Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
| | - Wenzhong Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (W.W.); (S.C.); (L.Q.); (S.Z.); (K.Y.); (Y.P.); (J.L.)
- Key Laboratory of Farm Animal Genetic Resources Exploration and Breeding of Shanxi Province, Jinzhong 030801, China
| |
Collapse
|
6
|
Glimpses from My Academic Journey : Based on the 15th Dr. K. C. Chaudhuri Lifetime Achievement Award Oration Delivered on 9th October 2022. Indian J Pediatr 2023; 90:69-75. [PMID: 36441386 DOI: 10.1007/s12098-022-04395-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
Abstract
This article is based on the contents of 'Dr. K. C. Chaudhuri Lifetime Achievement Award Oration' delivered on the Indian Journal of Pediatrics Annual Day 2022. The author shares glimpses of his academic journey from a remote village to a central Institute. This includes his career as a medical teacher and developing the Department of Neonatology at JIPMER, Pondicherry. This article is primarily focused on some of the significant research conducted during his tenure, like perinatal asphyxia, therapeutic hypothermia, neonatal sepsis, intrauterine growth restriction, and human milk banking.
Collapse
|
7
|
Zhou H, Zhao C, Wang P, Yang W, Zhu H, Zhang S. Regulators involved in trophoblast syncytialization in the placenta of intrauterine growth restriction. Front Endocrinol (Lausanne) 2023; 14:1107182. [PMID: 36798658 PMCID: PMC9927020 DOI: 10.3389/fendo.2023.1107182] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Placental dysfunction refers to the insufficiency of placental perfusion and chronic hypoxia during early pregnancy, which impairs placental function and causes inadequate supply of oxygen and nutrients to the fetus, affecting fetal development and health. Fetal intrauterine growth restriction, one of the most common outcomes of pregnancy-induced hypertensions, can be caused by placental dysfunction, resulting from deficient trophoblast syncytialization, inadequate trophoblast invasion and impaired vascular remodeling. During placental development, cytotrophoblasts fuse to form a multinucleated syncytia barrier, which supplies oxygen and nutrients to meet the metabolic demands for fetal growth. A reduction in the cell fusion index and the number of nuclei in the syncytiotrophoblast are found in the placentas of pregnancies complicated by IUGR, suggesting that the occurrence of IUGR may be related to inadequate trophoblast syncytialization. During the multiple processes of trophoblasts syncytialization, specific proteins and several signaling pathways are involved in coordinating these events and regulating placental function. In addition, epigenetic modifications, cell metabolism, senescence, and autophagy are also involved. Study findings have indicated several abnormally expressed syncytialization-related proteins and signaling pathways in the placentas of pregnancies complicated by IUGR, suggesting that these elements may play a crucial role in the occurrence of IUGR. In this review, we discuss the regulators of trophoblast syncytialization and their abnormal expression in the placentas of pregnancies complicated by IUGR.
Collapse
Affiliation(s)
- Hanjing Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Chenqiong Zhao
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Peixin Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Haiyan Zhu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| |
Collapse
|
8
|
Modulation of Nod-like Receptor Expression in the Thymus during Early Pregnancy in Ewes. Vaccines (Basel) 2022; 10:vaccines10122128. [PMID: 36560538 PMCID: PMC9781860 DOI: 10.3390/vaccines10122128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/24/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Nucleotide-binding oligomerization domain receptors (NOD-like receptors, NLRs) are involved in modulating the innate immune responses of the trophoblast and the placenta in normal pregnancy. The thymus participates in regulation of innate and adaptive immune responses. However, it is unclear whether expression of NLR is modulated in the maternal thymus during early pregnancy. In this study, thymuses were sampled at day 16 of the estrous cycle, and at days 13, 16 and 25 of gestation (n = 6 for each group) from ewes after slaughter. Different stages were chosen because the maternal thymus was under the different effects of interferon-tau and/or progesterone or not. RT-qPCR, Western blot and immunohistochemistry analysis were used to analyze the expression of the NLR family, including NOD1; NOD2; major histocompatibility complex class II transactivator (CIITA); NLR family apoptosis inhibitory protein (NAIP); nucleotide-binding oligomerization domain and Leucine-rich repeat and Pyrin domain containing protein 1 (NLRP1), NLRP3 and NLRP7. The results showed that expression level of NOD1 was changed with the pregnancy stages, and expression levels of NOD2, CIITA, NAIP, NLRP1, NLRP3 and NLRP7 mRNA and proteins were peaked at day 13 of pregnancy. The levels of NOD2 and CIITA were increased during early pregnancy. The stainings for NOD2 and NLRP7 proteins were located in epithelial reticular cells, capillaries and thymic corpuscles. In summary, pregnancy stages changed expression of NLR family in the maternal thymus, which may be related to the modulation of maternal thymic immune responses, and beneficial for normal pregnancy in sheep.
Collapse
|
9
|
Zhao Z, Li Y, Cao J, Fang H, Zhang L, Yang L. Early Pregnancy Modulates Expression of the Nod-like Receptor Family in Lymph Nodes of Ewes. Animals (Basel) 2022; 12:ani12233285. [PMID: 36496806 PMCID: PMC9738492 DOI: 10.3390/ani12233285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
NOD receptors (NLRs) mediate adaptive immune responses and immune tolerance. Nevertheless, it is not clear if gestation modulates the NLR signaling pathway in lymph nodes of ewes. In this study, lymph nodes of ewes were collected at day 16 of the estrous cycle, and at days 13, 16 and 25 of gestation (n = 6 for each group). RT-qPCR, Western blot and immunohistochemistry analysis were used to analyze the expression of the NLR family, including NOD1, NOD2, CIITA, NAIP, NLRP1, NLRP3 and NLRP7. The data showed that early gestation enhanced expression of NOD1, CIITA, NLRP1, NLRP3 and NLRP7 mRNA, as well as proteins at day 16 of gestation, and the expression levels of NOD2, CIITA, NLRP1 and NLRP7 were higher at days 13 and 25 of gestation than day 16 of the estrous cycle. However, NOD1 expression was lower on days 13 and 25 of gestation compared to day 16 of the estrous cycle, and early gestation suppressed NAIP expression. In summary, early pregnancy modulated expression of the NLR family in ovine lymph nodes, which participates in immune regulation, and this modulation may be necessary for pregnancy establishment in ewes.
Collapse
|
10
|
Yang H, He J, Huang S, Yang H, Yi Q, Tao Y, Chen M, Zhang X, Qi H. Identification and Characterization of Extrachromosomal Circular DNA in Human Placentas With Fetal Growth Restriction. Front Immunol 2022; 12:780779. [PMID: 34992600 PMCID: PMC8724250 DOI: 10.3389/fimmu.2021.780779] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Many studies have confirmed that extrachromosomal circular DNAs (eccDNAs/ecDNAs) exist in tumor and normal cells independently of the chromosome and are essential for oncogene plasticity and drug resistance. Studies have confirmed that there are many eccDNAs/ecDNAs in maternal plasma derived from the fetus. Fetal growth restriction (FGR) is a pregnancy-related disease associated with high newborn morbidity and mortality. However, the characteristics and nature of eccDNAs/ecDNAs in FGR are poorly understood. This study aims to deconstruct the properties and potential functions of eccDNAs/ecDNAs in FGR. We performed circle-seq to identify the expression profile of eccDNAs/ecDNAs, analyzed by bioinformatics, and verified by real-time Polymerase Chain Reaction (PCR) combined with southern blot in FGR compared with the normal groups. A total of 45,131 eccDNAs/ecDNAs (including 2,118 unique ones) were identified, which had significantly higher abundance in FRG group than in normal group, and was bimodal in length, peaking at ~146bp and ~340bp, respectively. Gestational age may be one independent factor affecting the production of eccDNAs/ecDNAs, most of which come from genomic regions with high gene density, with a 4~12bp repeat around the junction, and their origin had a certain genetic preference. In addition, some of the host-genes overlapped with non-coding RNAs (ncRNAs) partially or even completely. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that host-genes on the differentially expressed eccDNAs/ecDNAs (DEEECs/DEECs) were mainly enriched in immune-related functions and pathways. The presence of some ecDNAs were verified, and whose variability were consistent with the circle-seq results. We identified and characterized eccDNAs/ecDNAs in placentas with FGR, and elucidated the formation mechanisms and the networks with ncRNAs, which provide a new vision for the screening of new biomarkers and therapeutic targets for FGR.
Collapse
Affiliation(s)
- Huan Yang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Obstetrics, Chongqing University Three Gorges Hospital, Chongqing, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,China-Canada-New Zealand Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jie He
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,China-Canada-New Zealand Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shuai Huang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,China-Canada-New Zealand Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Hongbing Yang
- Department of Obstetrics, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Qingjie Yi
- Department of Obstetrics, Chongqing University Three Gorges Hospital, Chongqing, China.,Department of Epidemiology and Health Statistics, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Yuelan Tao
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,China-Canada-New Zealand Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Miaomiao Chen
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,China-Canada-New Zealand Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,China-Canada-New Zealand Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China.,China-Canada-New Zealand Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Zhang C, Ding J, Li H, Wang T. Identification of key genes in pathogenesis of placental insufficiency intrauterine growth restriction. BMC Pregnancy Childbirth 2022; 22:77. [PMID: 35090410 PMCID: PMC8796578 DOI: 10.1186/s12884-022-04399-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/10/2022] [Indexed: 01/10/2023] Open
Abstract
Background Intrauterine growth restriction (IUGR) is defined as a fetus that fails to achieve its genetically determined growth potential. The exact molecular mechanisms of placental insufficiency IUGR pathogenesis are a little known. Our goal was to identify key genes and gene co-expression modules related to placental insufficiency IUGR. Methods We used weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) network analysis to examine the IUGR dataset GSE114691 from NCBI Gene Expression Omnibus. Core modules and hub nodes of the protein-protein interaction network were identified. A gene network was constructed and genes were classified by WGCNA into different modules. The validation of potential key genes was carried out using additional datasets (GSE12216 and GSE24129). Results We identified in GSE114691 539 down regulated genes and 751 up regulated genes in placental tissues characteristic of placental insufficiency IUGR compared with non-IUGR, and defined 76 genes as hub nodes in the protein-protein interaction network. Genes in the key modules of the WGCNA network were most closely associated with placental insufficiency IUGR and significantly enriched in biological process such as cellular metabolic process and macromolecule metabolic process. We identified as key genes TGFB1, LEP, ENG, ITGA5, STAT5A, LYN, GATA3, FPR1, TGFB2, CEBPB, KLF4, FLT1, and PNPLA2. The RNA expression levels of ENG and LEP, as biomarkers, were validated. Conclusion A holistic gene expression profile of placental insufficiency IUGR has been generated and the key genes ENG and LEP has potential to serve as circulating diagnosis biomarkers and therapeutic targets for placental insufficiency IUGR. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04399-3.
Collapse
|
12
|
Chen M, Ma Y, Ma T, Li Y, Gao D, Chen L, Liu J, Zhang Y, Jiang J, Wang X, Dong Y, Ma J. The association between growth patterns and blood pressure in children and adolescents: A cross-sectional study of seven provinces in China. J Clin Hypertens (Greenwich) 2021; 23:2053-2064. [PMID: 34847290 PMCID: PMC8696227 DOI: 10.1111/jch.14393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 01/22/2023]
Abstract
Aimed to investigate the associations between different growth patterns with high blood pressure, and further examine the mediation effect of BMI between growth patterns and high blood pressure among children and adolescents. A total of 31581 children and adolescents aged 7-18 years were selected based on the stratified cluster sampling method. Logistics regression models were used to calculate the odds rations (ORs) and 95% confidence interval (95%CI) of the association between different growth patterns and high blood pressure. Mediation effect analyses were applied to estimate the effect of BMI on the increase of blood pressure levels in different growth patterns. In different sex and ages, compared to reference group of normal growth, blood pressure levels and prevalence of high blood pressure of the catch-up growth were higher, but that of the catch-down growth were lower. The prevalence of high blood pressure was 11.69%, 16.06%, and 9.68% in normal growth, catch-up growth, and catch-down growth, respectively. In total, compared with the normal growth pattern, the ORs (95%CI) of high blood pressure, high systolic blood pressure and high diastolic blood pressure in the catch-up growth were 1.171(1.073,1.280), 1.110(1.001,1.230) and 1.141(1.025,1.270) (p < .05), respectively. Additionally, the mediation effect of current BMI existed in the association between blood pressure levels and different growth patterns, particularly in boys. Our findings suggested that different growth patterns after birth could modify blood pressure, and the potential risks of high blood pressure could be increased by catch-up growth at childhood and adolescence.
Collapse
Affiliation(s)
- Manman Chen
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Ying Ma
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Tao Ma
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Yanhui Li
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Di Gao
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Li Chen
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Jieyu Liu
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Yi Zhang
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Jun Jiang
- Department of Plant Science and Landscape ArchitectureUniversity of MarylandCollege ParkMarylandUSA
| | - Xinxin Wang
- School of Public Health and ManagementKey Laboratory of Environmental Factors and Chronic Disease ControlNingxia Medical UniversityNingxiaChina
| | - Yanhui Dong
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Jun Ma
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| |
Collapse
|
13
|
Huang L, Jiang S, Xu J, Lei X, Zhang J. Associations between prepregnancy body mass index, gestational weight gain and weight catch-up in small-for-gestational-age children. MATERNAL AND CHILD NUTRITION 2021; 18:e13235. [PMID: 34291873 PMCID: PMC8710114 DOI: 10.1111/mcn.13235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 06/05/2021] [Accepted: 06/16/2021] [Indexed: 11/29/2022]
Abstract
Inadequate gestational weight gain (GWG) was related with a higher incidence of small‐for‐gestational‐age (SGA) births than appropriate GWG; however, the long‐term association of maternal GWG with weight catch‐up growth in SGA children remains unknown. The objective of this study is to evaluate the associations between prepregnancy body mass index (pBMI), GWG and weight catch‐up patterns in SGA children. Data were from the Collaborative Perinatal Project, an American multicentre prospective cohort study. A total of 56,990 gravidas were recruited at the first prenatal visit, and children were followed up until school age. Maternal pBMI, GWG and physical growth of the offspring at birth, 4 months, 1 year, 4 years and 7 years old were recorded. The latent class analysis was employed to form weight catch‐up growth patterns (appropriate, excessive, slow, regression and no catch‐up patterns) in SGA children. SGA children who developed the ‘appropriate catch‐up growth’ pattern and whose mothers had appropriate pBMI and GWG were chosen as the reference. Associations between GWG for different pBMI and weight catch‐up patterns were analysed by multivariate logistic regression models. A total of 1619 infants (9.45%) were born term SGA. After adjusting for relevant confounders, compared with SGA children whose mothers had appropriate pBMI and GWG, SGA children with maternal prepregnancy underweight (for inadequate GWG, GWG below recommendations, adjusted OR: 2.88, 95% CI: 1.13–7.31; for appropriate/excessive GWG, adjusted OR: 3.07, 95% CI: 1.74–5.42) or with prepregnancy normal weight but inadequate GWG (adjusted OR: 2.14, 95% CI: 1.36–3.38) were at a higher risk of having the ‘no catch‐up growth’ pattern. We suggest that SGA children with maternal prepregnancy underweight or inadequate GWG tend to have a poor weight catch‐up growth at least until school age.
Collapse
Affiliation(s)
- Lihua Huang
- The International Peace Maternity & Child Health Hospital of China Welfare Institute, Department of Child Healthcare, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Xinhua Hospital, MOE-Shanghai Key Laboratory of Children's Environmental Health, Department of Child and Adolescent Healthcare, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiwei Jiang
- The International Peace Maternity & Child Health Hospital of China Welfare Institute, Department of Child Healthcare, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Xinhua Hospital, MOE-Shanghai Key Laboratory of Children's Environmental Health, Department of Child and Adolescent Healthcare, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Xu
- The International Peace Maternity & Child Health Hospital of China Welfare Institute, Department of Child Healthcare, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Xinhua Hospital, MOE-Shanghai Key Laboratory of Children's Environmental Health, Department of Child and Adolescent Healthcare, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Lei
- Xinhua Hospital, MOE-Shanghai Key Laboratory of Children's Environmental Health, Department of Child and Adolescent Healthcare, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Neonatology, Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Jun Zhang
- Xinhua Hospital, MOE-Shanghai Key Laboratory of Children's Environmental Health, Department of Child and Adolescent Healthcare, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Sanchez-Aranguren L, Nadeem S. Bioenergetics adaptations and redox homeostasis in pregnancy and related disorders. Mol Cell Biochem 2021; 476:4003-4018. [PMID: 34196872 PMCID: PMC8473347 DOI: 10.1007/s11010-021-04215-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Pregnancy is a challenging physiological process that involves maternal adaptations to the increasing energetics demands imposed by the growing conceptus. Failure to adapt to these requirements may result in serious health complications for the mother and the baby. The mitochondria are biosynthetic and energy-producing organelles supporting the augmented energetic demands of pregnancy. Evidence suggests that placental mitochondria display a dynamic phenotype through gestation. At early stages of pregnancy placental mitochondria are mainly responsible for the generation of metabolic intermediates and reactive oxygen species (ROS), while at later stages of gestation, the placental mitochondria exhibit high rates of oxygen consumption. This review describes the metabolic fingerprint of the placental mitochondria at different stages of pregnancy and summarises key signs of mitochondrial dysfunction in pathological pregnancy conditions, including preeclampsia, gestational diabetes and intrauterine growth restriction (IUGR). So far, the effects of placental-driven metabolic changes governing the metabolic adaptations occurring in different maternal tissues in both, healthy and pathological pregnancies, remain to be uncovered. Understanding the function and molecular aspects of the adaptations occurring in placental and maternal tissue's mitochondria will unveil potential targets for further therapeutic exploration that could address pregnancy-related disorders. Targeting mitochondrial metabolism is an emerging approach for regulating mitochondrial bioenergetics. This review will also describe the potential therapeutic use of compounds with a recognised effect on mitochondria, for the management of preeclampsia.
Collapse
Affiliation(s)
| | - Sarah Nadeem
- College of Health and Life Sciences, Aston Medical School, Aston University, Birmingham, UK
| |
Collapse
|
15
|
Kuiper-Makris C, Selle J, Nüsken E, Dötsch J, Alejandre Alcazar MA. Perinatal Nutritional and Metabolic Pathways: Early Origins of Chronic Lung Diseases. Front Med (Lausanne) 2021; 8:667315. [PMID: 34211985 PMCID: PMC8239134 DOI: 10.3389/fmed.2021.667315] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Lung development is not completed at birth, but expands beyond infancy, rendering the lung highly susceptible to injury. Exposure to various influences during a critical window of organ growth can interfere with the finely-tuned process of development and induce pathological processes with aberrant alveolarization and long-term structural and functional sequelae. This concept of developmental origins of chronic disease has been coined as perinatal programming. Some adverse perinatal factors, including prematurity along with respiratory support, are well-recognized to induce bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease that is characterized by arrest of alveolar and microvascular formation as well as lung matrix remodeling. While the pathogenesis of various experimental models focus on oxygen toxicity, mechanical ventilation and inflammation, the role of nutrition before and after birth remain poorly investigated. There is accumulating clinical and experimental evidence that intrauterine growth restriction (IUGR) as a consequence of limited nutritive supply due to placental insufficiency or maternal malnutrition is a major risk factor for BPD and impaired lung function later in life. In contrast, a surplus of nutrition with perinatal maternal obesity, accelerated postnatal weight gain and early childhood obesity is associated with wheezing and adverse clinical course of chronic lung diseases, such as asthma. While the link between perinatal nutrition and lung health has been described, the underlying mechanisms remain poorly understood. There are initial data showing that inflammatory and nutrient sensing processes are involved in programming of alveolarization, pulmonary angiogenesis, and composition of extracellular matrix. Here, we provide a comprehensive overview of the current knowledge regarding the impact of perinatal metabolism and nutrition on the lung and beyond the cardiopulmonary system as well as possible mechanisms determining the individual susceptibility to CLD early in life. We aim to emphasize the importance of unraveling the mechanisms of perinatal metabolic programming to develop novel preventive and therapeutic avenues.
Collapse
Affiliation(s)
- Celien Kuiper-Makris
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Nüsken
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A. Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Member of the German Centre for Lung Research (DZL), Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Gießen, Germany
| |
Collapse
|
16
|
de Knegt VE, Hedley PL, Kanters JK, Thagaard IN, Krebs L, Christiansen M, Lausten-Thomsen U. The Role of Leptin in Fetal Growth during Pre-Eclampsia. Int J Mol Sci 2021; 22:ijms22094569. [PMID: 33925454 PMCID: PMC8123779 DOI: 10.3390/ijms22094569] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Leptin is secreted by the placenta and has a multi-facetted role in the regulation of functions related to pregnancy. Metabolic disorders and insufficient homeostatic compensatory mechanisms involving leptin during pregnancy play a decisive role in the development of pre-eclampsia (PE) and give rise to compromised intrauterine growth conditions and aberrant birth weight of offspring. This review was compiled to elucidate the metabolic background of PE and its relationship with adverse intrauterine growth conditions through the examination of leptin as well as to describe possible mechanisms linking leptin to fetal growth restriction. This review illustrates that leptin in PE is dysregulated in maternal, fetal, and placental compartments. There is no single set of unifying mechanisms within the spectrum of PE, and regulatory mechanisms involving leptin are specific to each situation. We conclude that dysregulated leptin is involved in fetal growth at many levels through complex interactions with parallel pregnancy systems and probably throughout the entirety of pregnancy.
Collapse
Affiliation(s)
- Victoria E. de Knegt
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institute, Artillerivej 5, 2300 Copenhagen, Denmark; (P.L.H.); (M.C.)
- Correspondence: ; Tel.: +45-50469429
| | - Paula L. Hedley
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institute, Artillerivej 5, 2300 Copenhagen, Denmark; (P.L.H.); (M.C.)
| | - Jørgen K. Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Science, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
| | - Ida N. Thagaard
- Department of Gynecology and Obstetrics, Copenhagen University Hospital Slagelse, Ingemannsvej 18, 4200 Slagelse, Denmark;
| | - Lone Krebs
- Department of Obstetrics and Gynecology, University Hospital Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark;
| | - Michael Christiansen
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institute, Artillerivej 5, 2300 Copenhagen, Denmark; (P.L.H.); (M.C.)
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Ulrik Lausten-Thomsen
- Department of Neonatology, University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark;
| |
Collapse
|
17
|
Intrauterine growth restriction: Clinical consequences on health and disease at adulthood. Reprod Toxicol 2021; 99:168-176. [DOI: 10.1016/j.reprotox.2020.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
|
18
|
Fantone S, Mazzucchelli R, Giannubilo SR, Ciavattini A, Marzioni D, Tossetta G. AT-rich interactive domain 1A protein expression in normal and pathological pregnancies complicated by preeclampsia. Histochem Cell Biol 2020; 154:339-346. [PMID: 32529396 DOI: 10.1007/s00418-020-01892-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/24/2022]
Abstract
AT-rich interactive domain 1A (ARID1A, as known as BAF250a) is a subunit of human switch/sucrose nonfermentable chromatin remodeling complex with tumour suppressor function. Mutations of Arid1a have been reported in many human cancers and low expression of this protein has been correlated to a poor prognosis outcome in patients affected by some types of cancer. Although there are many studies regarding ARID1A functions in cancer, little is known about its role in regulating cell differentiation and normal tissues homeostasis. Here, we investigate ARID1A expression in normal placental tissues of first and third trimester of gestation and in pathological placental tissues of pregnancy complicated by preeclampsia (PE) and intrauterine growth restriction (IUGR) to evaluate a possible role of this protein in trophoblast differentiation. We found that ARID1A was specifically expressed in villous and extravillous cytotrophoblastic cells in normal placentas whereas syncytiotrophoblast was negative. Interestingly, ARID1A was expressed in both cytotrophoblastic cells and syncytiotrophoblast in placentas affected by PE and PE-IUGR. Moreover, ARID1A was also present in syncitial knots of pathological placentas. The present results indicate that ARID1A is a good marker of poor trophoblast differentiation in these pathologies, because the significant high positive staining in syncytiotrophoblast nuclei may suggest a poor differentiation of this trophoblast layer due to the cytotrophoblast cells fusion with the syncytiotrophoblast overlaying before arresting their cell cycle.
Collapse
Affiliation(s)
- Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Roberta Mazzucchelli
- Section of Pathological Anatomy, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, United Hospitals, Ancona, Italy
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy
| | - Andrea Ciavattini
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.,Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy
| |
Collapse
|
19
|
de Aguiar Greca SC, Kyrou I, Pink R, Randeva H, Grammatopoulos D, Silva E, Karteris E. Involvement of the Endocrine-Disrupting Chemical Bisphenol A (BPA) in Human Placentation. J Clin Med 2020; 9:jcm9020405. [PMID: 32028606 PMCID: PMC7074564 DOI: 10.3390/jcm9020405] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Endocrine-disrupting chemicals (EDCs) are environmental chemicals/toxicants that humans are exposed to, interfering with the action of multiple hormones. Bisphenol A (BPA) is classified as an EDC with xenoestrogenic activity with potentially adverse effects in reproduction. Currently, a significant knowledge gap remains regarding the complete spectrum of BPA-induced effects on the human placenta. As such, the present study examined the effects of physiologically relevant doses of BPA in vitro. Methods: qRT-PCR, Western blotting, immunofluorescence, ELISA, microarray analyses, and bioinformatics have been employed to study the effects of BPA using nonsyncytialised (non-ST) and syncytialised (ST) BeWo cells. Results: Treatment with 3 nM BPA led to an increase in cell number and altered the phosphorylation status of p38, an effect mediated primarily via the membrane-bound estrogen receptor (GPR30). Nonbiased microarray analysis identified 1195 and 477 genes that were differentially regulated in non-ST BeWo cells, whereas in ST BeWo cells, 309 and 158 genes had altered expression when treated with 3 and 10 nM, respectively. Enriched pathway analyses in non-ST BeWo identified a leptin and insulin overlap (3 nM), methylation pathways (10 nM), and differentiation of white and brown adipocytes (common). In the ST model, most significantly enriched were the nuclear factor erythroid 2-related factor 2 (NRF2) pathway (3 nM) and mir-124 predicted interactions with cell cycle and differentiation (10 nM). Conclusion: Collectively, our data offer a new insight regarding BPA effects at the placental level, and provide a potential link with metabolic changes that can have an impact on the developing fetus.
Collapse
Affiliation(s)
| | - Ioannis Kyrou
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK;
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute of Precision Diagnostics and Translational Medicine, UHCW NHS Trust, Coventry CV4 7AL, UK; (H.R.); (D.G.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Ryan Pink
- Dept of Bio. & Med. Sci., Oxford Brookes University, Oxford OX3 0BP, UK;
| | - Harpal Randeva
- Institute of Precision Diagnostics and Translational Medicine, UHCW NHS Trust, Coventry CV4 7AL, UK; (H.R.); (D.G.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Dimitris Grammatopoulos
- Institute of Precision Diagnostics and Translational Medicine, UHCW NHS Trust, Coventry CV4 7AL, UK; (H.R.); (D.G.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Elisabete Silva
- College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK;
- Correspondence: (E.S.); (E.K.)
| | - Emmanouil Karteris
- College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK;
- Correspondence: (E.S.); (E.K.)
| |
Collapse
|
20
|
Gomez SD, Bustos PS, Sánchez VG, Ortega MG, Guiñazú N. Trophoblast toxicity of the neonicotinoid insecticide acetamiprid and an acetamiprid-based formulation. Toxicology 2020; 431:152363. [DOI: 10.1016/j.tox.2020.152363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/11/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
|
21
|
Sonkar R, Kay MK, Choudhury M. PFOS Modulates Interactive Epigenetic Regulation in First-Trimester Human Trophoblast Cell Line HTR-8/SV neo. Chem Res Toxicol 2019; 32:2016-2027. [PMID: 31508952 DOI: 10.1021/acs.chemrestox.9b00198] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Organic compounds have been linked to adverse pregnancy complications. Perfluorooctanesulfonic acid (PFOS), a man-made fluorosurfactant and global pollutant, has been shown to induce oxidative stress in various cell types. Oxidative stress plays a key role in leading several placental diseases including preeclampsia (PE), gestational diabetes, spontaneous abortion, preterm labor, and intrauterine growth restriction. Recently, epigenetic regulation such as histone modifications, DNA methylation, and microRNAs (miRNAs), are shown to be associated with oxidative stress as well as pregnancy complications such as PE. However, whether PFOS exerts its detrimental effects in the placenta through epigenetics remains to be unveiled. Therefore, we aimed to investigate the effect of PFOS-induced reactive oxygen species (ROS) generation in first trimester human trophoblast cell line (HTR-8/SVneo) and whether epigenetic regulation is involved in this process. When treated with a range of PFOS doses at 24 and 48 h, even at 10 μM, it significantly increased the ROS production and decreased gene and protein expression, respectively, of the DNA methyltransferases DNMT1 (p < 0.001; p < 0.05), DNMT3A (p < 0.001; p < 0.05), and DNMT3B (p < 0.01; p < 0.01) and the sirtuins, for example, SIRT1 (p < 0.001; p < 0.001) and SIRT3 (p < 0.001; p < 0.05), while reducing global DNA methylation (p < 0.01) and increasing protein lysine acetylation (p < 0.001) as compared to vehicle controls. Interestingly, PFOS (10 μM) significantly increased miR29-b (p < 0.01), which has been previously reported to be associated with PE. The observed epigenetic effects were shown to be dependent on the expression of miR-29b, as knockdown of miR-29b significantly alters the gene and protein expression of DNMT1, DNMT3A, DNMT3B, SIRT1, and SIRT3 and ROS production as well as global DNA methylation and protein acetylation. This study provides for the first time a novel insight into PFOS-induced ROS generation via regulation of sets of the interactive epigenetic circuit in the placenta, which may lead to pregnancy complications.
Collapse
Affiliation(s)
- Ravi Sonkar
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , 312 REYN, MS 1114 , College Station , Texas 77843 , United States
| | - Matthew K Kay
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , 312 REYN, MS 1114 , College Station , Texas 77843 , United States
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , 312 REYN, MS 1114 , College Station , Texas 77843 , United States
| |
Collapse
|
22
|
Quintero-Ronderos P, Jiménez KM, Esteban-Pérez C, Ojeda DA, Bello S, Fonseca DJ, Coronel MA, Moreno-Ortiz H, Sierra-Díaz DC, Lucena E, Barbaux S, Vaiman D, Laissue P. FOXD1 mutations are related to repeated implantation failure, intra-uterine growth restriction and preeclampsia. Mol Med 2019; 25:37. [PMID: 31395028 PMCID: PMC6688323 DOI: 10.1186/s10020-019-0104-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
Background Human reproductive disorders consist of frequently occurring dysfunctions including a broad range of phenotypes affecting fertility and women’s health during pregnancy. Several female-related diseases have been associated with hypofertility/infertility phenotypes, such as recurrent pregnancy loss (RPL). Other occurring diseases may be life-threatening for the mother and foetus, such as preeclampsia (PE) and intra-uterine growth restriction (IUGR). FOXD1 was defined as a major molecule involved in embryo implantation in mice and humans by regulating endometrial/placental genes. FOXD1 mutations in human species have been functionally linked to RPL’s origin. Methods FOXD1 gene mutation screening, in 158 patients affected by PE, IUGR, RPL and repeated implantation failure (RIF), by direct sequencing and bioinformatics analysis. Plasmid constructs including FOXD1 mutations were used to perform in vitro gene reporter assays. Results Nine non-synonymous sequence variants were identified. Functional experiments revealed that p.His267Tyr and p.Arg57del led to disturbances of promoter transcriptional activity (C3 and PlGF genes). The FOXD1 p.Ala356Gly and p.Ile364Met deleterious mutations (previously found in RPL patients) have been identified in the present work in women suffering PE and IUGR. Conclusions Our results argue in favour of FOXD1 mutations’ central role in RPL, RIF, IUGR and PE pathogenesis via C3 and PlGF regulation and they describe, for the first time, a functional link between FOXD1 and implantation/placental diseases. FOXD1 could therefore be used in clinical environments as a molecular biomarker for these diseases in the near future. Keywords Recurrent pregnancy loss, Preeclampsia, Intra-uterine growth restriction, FOXD1 Electronic supplementary material The online version of this article (10.1186/s10020-019-0104-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paula Quintero-Ronderos
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Karen Marcela Jiménez
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Clara Esteban-Pérez
- Fertility and Sterility Colombian Center, Department of Reproductive Genetics, Bogotá, Colombia
| | - Diego A Ojeda
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.,Clinical Neurosciences and Psychiatry, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK., Southampton, United Kingdom
| | - Sandra Bello
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Dora Janeth Fonseca
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - María Alejandra Coronel
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Harold Moreno-Ortiz
- Fertility and Sterility Colombian Center, Department of Reproductive Genetics, Bogotá, Colombia
| | - Diana Carolina Sierra-Díaz
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Elkin Lucena
- Fertility and Sterility Colombian Center, Department of Reproductive Genetics, Bogotá, Colombia
| | - Sandrine Barbaux
- Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France
| | - Daniel Vaiman
- Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France
| | - Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
23
|
Nicotine protects fetus against LPS-induced fetal growth restriction through ameliorating placental inflammation and vascular development in late pregnancy in rats. Biosci Rep 2019; 39:BSR20190386. [PMID: 31209145 PMCID: PMC6603276 DOI: 10.1042/bsr20190386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/14/2019] [Accepted: 06/12/2019] [Indexed: 01/19/2023] Open
Abstract
Our previous work has shown that nicotine suppressed lipopolysaccharide (LPS)-induced placental inflammation by inhibiting cytokine release as well as infiltration of leukocytes into the placenta through the cholinergic anti-inflammatory pathway. Nicotine also increased fetal survival and restored pup weight. In the present study, we aim to further investigate if fetal growth restriction (FGR) occurs with LPS treatment, and evaluate the protective effects of nicotine on fetuses in late gestation of rats. Pregnant Sprague–Dawley rats were divided into control group, nicotine group, LPS group and LPS + nicotine group. Rats were first pretreated with nicotine or vehicle by subcutaneous injection on gestation day (GD)14 and GD15, followed by LPS or vehicle intraperitoneal injection on GD16, and were killed on GD18. Loss of fetuses, number and weights of live fetuses and weights of placentas were recorded. Placentas were collected to evaluate placental pathology and determine inflammatory cytokines and vascular endothelial growth factor (VEGF) levels. We found that LPS treatment increased levels of placental inflammatory cytokines and placental pathological damage, decreased levels of VEGF, reduced number of live fetuses and induced FGR. Pretreatment with nicotine reversed LPS-induced high levels of placental inflammatory cytokines, low levels of placental VEGF and placental pathological damage, then rescued the number and weights of live fetuses. These data demonstrated that activation of the cholinergic anti-inflammatory pathway by nicotine protected fetus against LPS-induced FGR through ameliorating placental inflammation and vascular development in late pregnancy in rats. It may be an alternative therapeutic strategy for inflammation- induced FGR in late pregnancy.
Collapse
|
24
|
Skoll A, Boutin A, Bujold E, Burrows J, Crane J, Geary M, Jain V, Lacaze-Masmonteil T, Liauw J, Mundle W, Murphy K, Wong S, Joseph KS. N° 364 - La Corticothérapie Prénatale Pour Améliorer Les Issues Néonatales. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2018; 40:1240-1262. [PMID: 30268317 DOI: 10.1016/j.jogc.2018.06.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIF Évaluer les avantages et les risques de la corticothérapie prénatale chez les femmes qui présentent un risque d'accouchement prématuré ou qui subissent une césarienne à terme avant début de travail, et formuler des recommandations visant l'amélioration des issues néonatales et des issues à long terme. OPTIONS Administrer ou ne pas administrer une corticothérapie prénatale aux femmes qui présentent un risque élevé d'accouchement prématuré ou qui subissent une césarienne avant travail à terme. RéSULTATS: Morbidité périnatale, notamment le syndrome de détresse respiratoire, l'hémorragie intraventriculaire, la dysplasie bronchopulmonaire, l'infection, l'hypoglycémie, ainsi que les troubles de la croissance somatique et cérébrale et du neurodéveloppement; mortalité périnatale; et morbidité maternelle, notamment l'infection et la suppression surrénalienne. UTILISATEURS CIBLES Fournisseurs de soins de maternité, notamment les sages-femmes, les médecins de famille et les obstétriciens. POPULATION CIBLE Femmes enceintes. ÉVIDENCE: Nous avons interrogé les bases de données Medline, PubMed et Embase ainsi que la Bibliothèque Cochrane, de leur création au mois de septembre 2017. Nous nous sommes servis de Medical Subjet Headings (MeSH) et de mots clés en lien avec la grossesse, la prématurité, les corticostéroïdes ainsi que la mortalité et la morbidité périnatales et néonatales. Nous avons également consulté les déclarations d'organismes professionnels tels que les National Institutes of Health, l'American College of Obstetricians and Gynecologists, la Society for Maternal-Fetal Medicine, le Royal College of Obstetricians and Gynaecologists et la Société canadienne de pédiatrie pour obtenir des références additionnelles. Les essais cliniques randomisés évaluant la corticothérapie prénatale menés sur des femmes enceintes et les revues systématiques antérieures sur le sujet étaient admissibles, tout comme les données venant de revues systématiques d'études non expérimentales (études de cohorte). VALEURS La présente opinion de comité a été révisée et approuvée par le Comité de médecine fœto-maternelle de la SOGC, et approuvée par le Conseil de la SOGC. AVANTAGES, INCONVéNIENTS ET COûTS: L'administration d'une corticothérapie prénatale dans les sept jours précédant l'accouchement réduit significativement la morbidité et la mortalité périnatales associées à la naissance prématurée survenant entre 24+0 et 34+6 semaines de grossesse. Si la corticothérapie prénatale est administrée plus de sept jours avant l'accouchement ou après 34+6 semaines de grossesse, les effets indésirables peuvent surpasser les avantages. Les données probantes sur l'impact à long terme de la corticothérapie prénatale sont rares. Par ailleurs, les effets neurodéveloppementaux néfastes potentiels de l'exposition répétée à la corticothérapie prénatale ou de l'administration de corticostéroïdes en période préterme tardive ou à terme n'ont pas été quantifiés. MIS-à-JOUR à LA DIRECTIVE: Une revue des données probantes sera menée cinq ans après la publication de la présente directive clinique afin d'évaluer si une mise à jour complète ou partielle s'impose. Si de nouvelles données probantes importantes sont publiées avant la fin de ces cinq ans, une mise à jour tenant compte des nouvelles connaissances et recommandations sera publiée. COMMANDITAIRES La présente directive clinique a été élaborée à l'aide de ressources fournies par la Société des obstétriciens et gynécologues du Canada et avec l'appui des Instituts de recherche en santé du Canada (APR-126338). MOTS CLéS: Corticothérapie prénatale, maturation fœtale, prématurité, période préterme tardive, césarienne avant travail DÉCLARATION SOMMAIRES: RECOMMANDATIONS: Considérations relatives à l'âge gestationnel.
Collapse
|