Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.960
Filter
1.
Rev Assoc Med Bras (1992) ; 70(9): e20231638, 2024.
Article in English | MEDLINE | ID: mdl-39292071

ABSTRACT

OBJECTIVE: Ghrelin is an adipokine the placenta generates to control the maternal metabolic adaptation to pregnancy. It causes different pregnancy complications like preeclampsia (PE). Therefore, the aim of this study was to assess the association between ghrelin mRNA expression and rs26311 and rs27647 polymorphisms and PE development. METHODS: In total, 156 PE women (including 97 patients with mild PE and 59 patients with severe PE) and 152 healthy controls were recruited in this case-control study during 2019-2020. All participants with other diseases have been excluded from both groups. The ghrelin expression was analyzed with real-time PCR, and ghrelin variants were examined using the RFLP-PCR method. RESULTS: The maternal and placental ghrelin rs27647 and rs26311 variants were unrelated to PE susceptibility. Haplotype analyses showed no significant difference between the four haplotypes and PE. No relationship was revealed between rs27647 polymorphism and severe PE. However, the results indicated a relationship between rs27647 and severe PE compared to mild PE and controls. Therefore, the rs27647 variant was associated with severe PE compared to mild PE in codominant, recessive, and log-additive models and controls in codominant, dominant, recessive, and log-additive models. The placental ghrelin mRNA expression declined in PE women compared to controls (0.67-fold), but the difference was insignificant (p=0.263). No significant difference was found between various genotypes of rs27647 and rs26311 polymorphisms concerning ghrelin mRNA expression. CONCLUSION: The maternal and placental ghrelin polymorphisms, rs27647 and rs26311, showed no effect on PE. However, the rs27647 variant was associated with severe PE.


Subject(s)
Genetic Predisposition to Disease , Ghrelin , Polymorphism, Single Nucleotide , Pre-Eclampsia , RNA, Messenger , Severity of Illness Index , Humans , Female , Pre-Eclampsia/genetics , Ghrelin/genetics , Pregnancy , Case-Control Studies , Adult , Genetic Predisposition to Disease/genetics , RNA, Messenger/genetics , Haplotypes , Genotype , Real-Time Polymerase Chain Reaction , Young Adult
2.
Hypertens Pregnancy ; 43(1): 2405857, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39316794

ABSTRACT

BACKGROUND: Preeclampsia and eclampsia are severe pregnancy disorders marked by hypertension and potential organ damage. The etiological basis of preeclampsia and eclampsia is not fully understood. Previous studies have revealed a link between sleep abnormality and preeclampsia/eclampsia, but the causal relationship remains unclear. In this study, we explored the genetic links between sleep and preeclampsia/eclampsia using genome-wide association study (GWAS) summary data and Mendelian randomization (MR) analysis. METHODS: RNA sequence dataset GSE114691 was downloaded from the Gene Expression Omnibus database, comprising placental tissues from patients with preeclampsia and controls. Differential expression analysis was conducted with R (v4.2.3) and DESeq2 (v1.38.3). Gene set enrichment analysis (GSEA) was carried out using HTSanalyzeR2. GWAS summary data on preeclampsia/eclampsia and genetic markers for sleep abnormality were sourced from the FinnGen Consortium and IEU genetic databases. The Mendelian randomization analysis was conducted with TwoSampleMR (v0.6.2), and the inverse variance weighted (IVW) approach was employed as the principal method. RESULTS: GSEA analysis revealed that the orexin receptor pathway showed heightened expression in the preeclampsia group versus controls. The random-effects IVW results showed that sleeplessness/insomnia has a genetic causal relationship with preeclampsia (OR = 2.08, 95% CI: 1.07-4.06, p = 0.0318), while sleep duration has evidence of regulating eclampsia (OR = 0.09, 95% CI: 0.01-0.67, p = 0.0187). CONCLUSION: This study provides significant evidence for a genetic causal association between sleep abnormalities and preeclampsia/eclampsia. [Figure: see text].


Subject(s)
Eclampsia , Genome-Wide Association Study , Mendelian Randomization Analysis , Pre-Eclampsia , Humans , Female , Pre-Eclampsia/genetics , Pregnancy , Eclampsia/genetics , Sleep Wake Disorders/genetics
3.
Front Endocrinol (Lausanne) ; 15: 1453277, 2024.
Article in English | MEDLINE | ID: mdl-39286274

ABSTRACT

Purpose: Our study utilizes Mendelian Randomization (MR) to explore the causal relationships between a range of risk factors and preeclampsia, a major contributor to maternal and perinatal morbidity and mortality. Methods: Employing the Inverse Variance Weighting (IVW) approach, we conducted a comprehensive multi-exposure MR study analyzing genetic variants linked to 25 risk factors including metabolic disorders, circulating lipid levels, immune and inflammatory responses, lifestyle choices, and bone metabolism. We applied rigorous statistical techniques such as sensitivity analyses, Cochran's Q test, MR Egger regression, funnel plots, and leave-one-out sensitivity analysis to address potential biases like pleiotropy and population stratification. Results: Our analysis included 267,242 individuals, focusing on European ancestries and involving 2,355 patients with preeclampsia. We identified strong genetic associations linking increased preeclampsia risk with factors such as hyperthyroidism, BMI, type 2 diabetes, and elevated serum uric acid levels. Conversely, no significant causal links were found with gestational diabetes, total cholesterol, sleep duration, and bone mineral density, suggesting areas for further investigation. A notable finding was the causal relationship between systemic lupus erythematosus and increased preeclampsia risk, highlighting the significant role of immune and inflammatory responses. Conclusion: This extensive MR study sheds light on the complex etiology of preeclampsia, underscoring the causal impact of specific metabolic, lipid, immune, lifestyle, and bone metabolism factors. Our findings advocate for a multidimensional approach to better understand and manage preeclampsia, paving the way for future research to develop targeted preventive and therapeutic strategies.


Subject(s)
Mendelian Randomization Analysis , Pre-Eclampsia , Humans , Female , Pregnancy , Pre-Eclampsia/genetics , Pre-Eclampsia/epidemiology , Risk Factors , White People/genetics , Europe/epidemiology , Genetic Predisposition to Disease , Adult , Polymorphism, Single Nucleotide
4.
J Matern Fetal Neonatal Med ; 37(1): 2389979, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39284760

ABSTRACT

OBJECTIVES: Pre-eclampsia (PE) and gestational hypertension (GH) are two different categories of hypertensive disorders of pregnancy. Given earlier observational research, the relationship between sex hormone-binding globulin (SHBG) and a higher risk of GH/PE is still up for dispute. Hence, the present investigation aimed to examine the possible link between SHBG and the likelihood of GH/PE. METHODS: As a first stage, single nucleotide polymorphisms from summary-level genome-wide association studies were tightly screened using quality-control techniques. Afterward, we utilized a two-sample Mendelian randomization (MR) study to examine the causal impact of SHBG on the likelihood of GH/PE. There was no indication of a relationship between blood SHBG level (n = 214,989) and GH/PE (1864 cases and 461,069 controls) in the initial study. Consensus results were obtained from the replicated analysis, which utilized MR estimates based on serum SHBG level(n = 214,989) for GH (4255 cases and 114,735 controls). RESULTS: The findings did not indicate any proof of a cause-and-effect connection between SHBG and the likelihood of GH/PE (odds ratio [OR] = 0.99, 95% confidence interval [CI] = 0.999 - 1.00, p = .34). Replicate analysis also revealed similar patterns (OR = 0.92, 95%CI = 0.82-1.05, p = .21). The above findings were demonstrated to have a strong level of robustness. CONCLUSIONS: The findings of this research did not offer definitive proof to endorse the idea that SHBG has a direct causal impact on the likelihood of GH/PE, which goes against numerous widely accepted observational studies. To ascertain the potential processes behind the relationships seen in observational studies, more investigation is needed.


Subject(s)
Genome-Wide Association Study , Hypertension, Pregnancy-Induced , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Pre-Eclampsia , Sex Hormone-Binding Globulin , Humans , Female , Sex Hormone-Binding Globulin/analysis , Pregnancy , Pre-Eclampsia/genetics , Pre-Eclampsia/blood , Pre-Eclampsia/epidemiology , Hypertension, Pregnancy-Induced/genetics , Hypertension, Pregnancy-Induced/blood , Hypertension, Pregnancy-Induced/epidemiology , Case-Control Studies
5.
Funct Integr Genomics ; 24(5): 157, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39237822

ABSTRACT

Aberrant long non-coding RNA (lncRNA) expression has been shown to be involved in the pathological process of pre-eclampsia (PE), yet only a small portion of lncRNAs has been characterized concerning the function and molecular mechanisms involved in PE. This study aimed to investigate the regulatory mechanism of the lncRNA AC092100.1 (AC092100.1) in angiogenesis in PE. In our study, bioinformatics analysis was performed to screen for differentially expressed lncRNAs between normal subjects and PE patients. The levels of AC092100.1 in placental tissues of patients with or without PE were validated using qRT-PCR. The effect of AC092100.1 overexpression on the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) was investigated. The binding of AC092100.1 and YT521-B homology domain-containing 2 (YTHDC2) was predicted and verified. The effect of AC092100.1/YTHDC2 on the expression of vascular endothelial growth factor-A (VEGFA) in HUVECs was determined. Finally, a PE mice model was conducted. Fetal mouse growth, the abundance of mesenchymal morphology markers, including hypoxia-inducible factor 1-alpha (HIF-1α), soluble fms-like tyrosine kinase-1 (sFlt-1), soluble endoglin (sEng), Slug, and Vimentin, and endothelial markers, including placental growth factor (PLGF), CD31, and vascular endothelial (VE)-cadherin, in placental tissues were assessed. Here, we found that AC092100.1 was abnormally downregulated in placental tissues from PE patients. We established that AC092100.1 overexpression promoted HUVEC proliferation, migration, and tube formation in vitro. Mechanistically, AC092100.1 induced the accumulation of YTHDC2 and VEGFA through binding to YTHDC2 in HUVECs. Inhibition of YTHDC2 or VEGFA reversed AC092100.1-promoted tube formation. AC092100.1 overexpression contributed to alleviating fetal growth disorder, decreased levels of sEng, HIF-1α, sFlt-1, Slug, and Vimentin, and increased levels of VEGFA, PLGF, CD31, and VE-cadherin in PE mice. Our findings provided evidence supporting the role of the AC092100.1/YTHDC2/VEGFA axis in regulating angiogenesis, which demonstrated a therapeutic pathway for PE targeting angiogenesis.


Subject(s)
Human Umbilical Vein Endothelial Cells , Pre-Eclampsia , RNA, Long Noncoding , Signal Transduction , Vascular Endothelial Growth Factor A , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/pathology , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Female , Pregnancy , Human Umbilical Vein Endothelial Cells/metabolism , Mice , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Proliferation , Cell Movement , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Placenta/metabolism , Angiogenesis
6.
Clin Epigenetics ; 16(1): 132, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294759

ABSTRACT

BACKGROUND: Imprinted genes play important functions in placentation and pregnancy; however, research on their roles in different placental diseases is limited. It is believed that epigenetic alterations, such as DNA methylation, of placental imprinting genes may contribute to the different pathological features of severe placental diseases, such as pre-eclampsia (PE) and placenta accreta spectrum disorders (PAS). RESULTS: In this study, we conducted a comparative analysis of the methylation and expression of placental imprinted genes between PE and PAS using bisulfite sequencing polymerase chain reaction (PCR) and quantitative PCR, respectively. Additionally, we assessed oxidative damage of placental DNA by determining 8-hydroxy-2'-deoxyguanosine levels and fetal growth by determining insulin-like growth factor 2 (IGF2) and cortisol levels in the umbilical cord blood using enzyme-linked immunosorbent assay. Our results indicated that methylation and expression of potassium voltage-gated channel subfamily Q member 1, GNAS complex locus, mesoderm specific transcript, and IGF2 were significantly altered in both PE and PAS placentas. Additionally, our results revealed that the maternal imprinted genes were significantly over-expressed in PE and significantly under-expressed in PAS compared with a normal pregnancy. Moreover, DNA oxidative damage was elevated and positively correlated with IGF2 DNA methylation in both PE and PAS placentas, and cortisol and IGF2 levels were significantly decreased in PE and PAS. CONCLUSIONS: This study suggested that DNA methylation and expression of imprinted genes are aberrant in both PE and PAS placentas and that PE and PAS have different methylation profiles, which may be linked to their unique pathogenesis.


Subject(s)
DNA Methylation , Genomic Imprinting , Insulin-Like Growth Factor II , Pre-Eclampsia , Humans , Female , Pregnancy , DNA Methylation/genetics , Genomic Imprinting/genetics , Insulin-Like Growth Factor II/genetics , Pre-Eclampsia/genetics , Adult , GTP-Binding Protein alpha Subunits, Gs/genetics , Placenta/metabolism , Epigenesis, Genetic/genetics , Hydrocortisone/blood , Placenta Diseases/genetics , Oxidative Stress/genetics , Fetal Blood/chemistry , Fetal Blood/metabolism , Chromogranins , Proteins , Potassium Channels, Voltage-Gated
7.
Int J Mol Sci ; 25(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273292

ABSTRACT

Preeclampsia (PE) is a major cause of maternal and neonatal morbidity and mortality worldwide, with the placenta playing a central role in disease pathophysiology. This review synthesizes recent advancements in understanding the molecular mechanisms underlying PE, focusing on placental genes, proteins, and genetic variants identified through multi-omic approaches. Transcriptomic studies in bulk placental tissue have identified many dysregulated genes in the PE placenta, including the PE signature gene, Fms-like tyrosine kinase 1 (FLT1). Emerging single-cell level transcriptomic data have revealed key cell types and molecular signatures implicated in placental dysfunction and PE. However, the considerable variability among studies underscores the need for standardized methodologies and larger sample sizes to enhance the reproducibility of results. Proteomic profiling of PE placentas has identified numerous PE-associated proteins, offering insights into potential biomarkers and pathways implicated in PE pathogenesis. Despite significant progress, challenges such as inconsistencies in study findings and lack of validation persist. Recent fetal genome-wide association studies have identified multiple genetic loci associated with PE, with ongoing efforts to elucidate their impact on placental gene expression and function. Future directions include the integration of multi-omic data, validation of findings in diverse PE populations and clinical subtypes, and the development of analytical approaches and experimental models to study the complex interplay of placental and maternal factors in PE etiology. These insights hold promise for improving risk prediction, diagnosis, and management of PE, ultimately reducing its burden on maternal and neonatal health.


Subject(s)
Placenta , Pre-Eclampsia , Proteomics , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Humans , Pregnancy , Female , Placenta/metabolism , Proteomics/methods , Genome-Wide Association Study , Transcriptome , Biomarkers , Multiomics
8.
Cell Mol Life Sci ; 81(1): 336, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120751

ABSTRACT

Preeclampsia (PE) is a life-threatening pregnancy-specific complication with controversial mechanisms and no effective treatment except delivery is available. Currently, increasing researchers suggested that PE shares pathophysiologic features with protein misfolding/aggregation disorders, such as Alzheimer disease (AD). Evidences have proposed defective autophagy as a potential source of protein aggregation in PE. Endoplasmic reticulum-selective autophagy (ER-phagy) plays a critical role in clearing misfolded proteins and maintaining ER homeostasis. However, its roles in the molecular pathology of PE remain unclear. We found that lncRNA DUXAP8 was upregulated in preeclamptic placentae and significantly correlated with clinical indicators. DUXAP8 specifically binds to PCBP2 and inhibits its ubiquitination-mediated degradation, and decreased levels of PCBP2 reversed the activation effect of DUXAP8 overexpression on AKT/mTOR signaling pathway. Function experiments showed that DUXAP8 overexpression inhibited trophoblastic proliferation, migration, and invasion of HTR-8/SVneo and JAR cells. Moreover, pathological accumulation of swollen and lytic ER (endoplasmic reticulum) was observed in DUXAP8-overexpressed HTR8/SVneo cells and PE placental villus trophoblast cells, which suggesting that ER clearance ability is impaired. Further studies found that DUXAP8 overexpression impaired ER-phagy and caused protein aggregation medicated by reduced FAM134B and LC3II expression (key proteins involved in ER-phagy) via activating AKT/mTOR signaling pathway. The increased level of FAM134B significantly reversed the inhibitory effect of DUXAP8 overexpression on the proliferation, migration, and invasion of trophoblasts. In vivo, DUXAP8 overexpression through tail vein injection of adenovirus induced PE-like phenotypes in pregnant rats accompanied with activated AKT/mTOR signaling, decreased expression of FAM134B and LC3-II proteins and increased protein aggregation in placental tissues. Our study reveals the important role of lncRNA DUXAP8 in regulating trophoblast biological behaviors through FAM134B-mediated ER-phagy, providing a new theoretical basis for understanding the pathogenesis of PE.


Subject(s)
Autophagy , Endoplasmic Reticulum , Pre-Eclampsia , Proto-Oncogene Proteins c-akt , RNA, Long Noncoding , Signal Transduction , TOR Serine-Threonine Kinases , Trophoblasts , Adult , Animals , Female , Humans , Pregnancy , Rats , Autophagy/genetics , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Endoplasmic Reticulum/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , TOR Serine-Threonine Kinases/metabolism , Trophoblasts/metabolism , Trophoblasts/pathology , Male
9.
Reprod Fertil Dev ; 362024 Aug.
Article in English | MEDLINE | ID: mdl-39094012

ABSTRACT

Context Preeclampsia is a common pregnancy complication, posing significant risks to both the mother and fetus. Predicting and determining the risks of this disease is crucial. Aims This research aims to understand the pathogenetic role of several factors in the development and progression of preeclampsia, particularly in relation to its severity in pregnant patients. Methods The study included 60 pregnant women diagnosed with either mild or severe preeclampsia and 40 healthy pregnant women for comparison. Blood plasma was analysed using biochemical methods, and blood microcirculation parameters were determined to identify homeostatic abnormalities in early preeclampsia. Key results A molecular genetic study revealed the frequency of the endothelial nitric oxide gene eNOSC774T . Homeostatic abnormalities were statistically correlated with polymorphic genotypes of the eNOSC774T gene. Conclusions The research found a correlation between the T774T eNOS genotype mutation and the severity of preeclampsia, alongside significant homeostasis abnormalities in patients. Implications The T774T mutant genotype of the eNOS gene and higher levels of lipid peroxidation products are strongly linked to the severity and progression of preeclampsia. This highlights a significant connection between genetic predisposition and biochemical abnormalities in the disease's development.


Subject(s)
Disease Progression , Nitric Oxide Synthase Type III , Pre-Eclampsia , Severity of Illness Index , Humans , Female , Pre-Eclampsia/genetics , Pregnancy , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Adult , Genetic Predisposition to Disease , Case-Control Studies , Genotype , Young Adult , Lipid Peroxidation/physiology , Mutation
11.
Mol Biol Rep ; 51(1): 895, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39115693

ABSTRACT

BACKGROUND: Insufficient trophoblast invasion, culminating in suboptimal uterine spiral artery remodeling, is pinpointed as a pivotal contributor to preeclampsia (PE) development. LINC01410 has been documented to be increased in various neoplasms, and is significantly associated with the invasive capabilities of tumor cells. Nonetheless, its function and the mechanisms in the pathogenesis of PE require further investigation. METHODS AND RESULTS: LINC01410 and methyltransferase-like 3 (METTL3) were ectopically expressed in HTR-8/Svneo cells via lentiviral transduction. Subsequently, the cells' invasive capabilities and apoptosis rates were evaluated employing Transwell assays and flow cytometry, respectively. The interplay between LINC01410 and METTL3, alongside the m6A methylation of FAS, was probed through RNA immunoprecipitation (RIP). Additionally, the association between FAS and METTL3 was elucidated via Coimmunoprecipitation (Co-IP) assays. The protein level of NF-κB, BAX, and BCL-2 in LINC01410-overexpressing cells was detected by Western blot. Our findings revealed that LINC01410 elevation increased the invasive ability of HTR-8/Svneo cells, directly impacting METTL3 then leading to its reduced expression. Conversely, heightened METTL3 expression mitigated invasiveness while enhancing apoptosis in these cells. Moreover, METTL3's interaction with FAS led to increased FAS expression, subject to m6A methylation. A surge in LINC01410 markedly decreased both mRNA and protein levels of FAS. Furthermore, LINC01410 overexpression significantly reduced NF-κB and BAX protein levels while augmenting BCL-2. CONCLUSIONS: Upregulation of LINC01410 expression promotes trophoblast cell invasion by inhibiting FAS levels through modified m6A alteration and suppressing the NF-κB pathway. These findings underscore the pivotal role of LINC01410 in regulating trophoblast cell invasion and propose it as a promising therapeutic strategy for preventing or alleviating PE. This offers valuable insights for the clinical treatment of PE, for which definitive targeted therapy methods are currently lacking.


Subject(s)
Apoptosis , Methyltransferases , Pre-Eclampsia , RNA, Long Noncoding , Trophoblasts , fas Receptor , Humans , Trophoblasts/metabolism , Methyltransferases/metabolism , Methyltransferases/genetics , fas Receptor/metabolism , fas Receptor/genetics , Female , Apoptosis/genetics , Pregnancy , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Cell Line , Cell Movement/genetics , NF-kappa B/metabolism , Signal Transduction/genetics
12.
BMC Pregnancy Childbirth ; 24(1): 543, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39148025

ABSTRACT

BACKGROUND: Preeclampsia is a severe obstetric disorder that significantly affects the maternal and neonatal peri-partum safety and long-term quality of life. However, there is limited research exploring the common mechanisms and potential clinical significance between early-onset preeclampsia and full-term preeclampsia from an immunological perspective. METHODS: In this study, data analysis was conducted. Initially, immune-related co-expressed genes involving both subtypes of preeclampsia were identified through Weighted Gene Co-expression Network Analysis (WGCNA). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were further employed to investigate the shared pathways regulated by immune-related genes. Binary logistic regression identified co-expressed genes with diagnostic value for preeclampsia, and a diagnostic model was constructed. Gene Set Enrichment Analysis (GSEA) predicted the potential biological functions of the selected genes. Lasso and Cox regression analyses identified genes closely associated with gestational duration, and a risk score model was established. A 4-gene feature, immune-related gene model for predicting the risk of preterm birth in preeclamptic pregnant women, was developed and validated through qPCR experiments. Immune cell infiltration analysis determined differences in immune cell infiltration between the two subtypes of preeclampsia. RESULTS: This study identified 4 immune-related co-expressed genes (CXCR6, PIK3CB, IL1RAP, and OSMR). Additionally, diagnostic and preterm birth risk prediction models for preeclampsia were constructed based on these genes. GSEA analysis suggested the involvement of these genes in the regulation of galactose metabolism, notch signaling pathway, and RIG-I like receptor signaling pathway. Immune pathway analysis indicated that the activation of T cell co-inhibition could be a potential intervention target for immunotherapy in early-onset preeclampsia. CONCLUSION: Our study provides promising insights into immunotherapy and mechanistic research for preeclampsia, discovering novel diagnostic and intervention biomarkers, and offering personalized diagnostic tools for preeclampsia.


Subject(s)
Pre-Eclampsia , Premature Birth , Adult , Female , Humans , Pregnancy , Clinical Relevance , Gene Expression Profiling , Gene Regulatory Networks , Pre-Eclampsia/diagnosis , Pre-Eclampsia/genetics , Pre-Eclampsia/immunology , Premature Birth/genetics , Premature Birth/immunology
13.
J Proteome Res ; 23(9): 4005-4013, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39171377

ABSTRACT

Ribosome profiling and mass spectrometry have revealed thousands of previously unannotated small and alternative open reading frames (sm/alt-ORFs) that are translated into micro/alt-proteins in mammalian cells. However, their prevalence across human tissues and biological roles remains largely undefined. The placenta is an ideal model for identifying unannotated microproteins and alt-proteins due to its considerable protein diversity that is required to sustain fetal development during pregnancy. Here, we profiled unannotated microproteins and alt-proteins in human placental tissues from preeclampsia patients or healthy individuals by proteomics, identified 52 unannotated microproteins or alt-proteins, and demonstrated that five microproteins can be translated from overexpression constructs in a heterologous cell line, although several are unstable. We further demonstrated that one microprotein, XRCC6P1, associates with translation initiation factor eIF3 and negatively regulates translation when exogenously overexpressed. Thus, we revealed a hidden sm/alt-ORF-encoded proteome in the human placenta, which may advance the mechanism studies for placenta development as well as placental disorders such as preeclampsia.


Subject(s)
Placenta , Pre-Eclampsia , Protein Biosynthesis , Proteomics , Humans , Pregnancy , Female , Placenta/metabolism , Proteomics/methods , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Open Reading Frames , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-3/genetics , Proteome/analysis , Proteome/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Micropeptides
14.
Int J Mol Sci ; 25(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39201555

ABSTRACT

Our incomplete knowledge of maternal-fetal interface (MFI) physiology impedes a better understanding of the pathological mechanisms leading to pregnancy complications, such as pre-eclampsia and fetal growth restriction. At the MFI, uterine natural killer (uNK) cells do not attack fetal cells but engage in crosstalk with both fetal and maternal cells to support feto-placental development. However, mother and fetus are genetically half-mismatched and certain combinations of variable immune genes-human leukocyte antigens (HLAs) and killer-cell immunoglobulin-like receptor (KIR), indeed, the most variable gene sets in the genome-associate with pregnancy outcomes, suggesting that these interactions regulate uNK cell function. How do these interactions influence the physiology and pathology at the MFI? Uterine NK cell function is regulated by both maternal and fetal Major Histocompatibility Complex (MHC); however, evidence for fetal cells educating uNK cells is lacking, and new evidence shows that maternal rather than fetal MHC class I molecules educate uNK cells. Furthermore, uNK cell education works through self-recognition by the ancient and conserved NKG2A receptor. Pregnant mice lacking this receptor produce normal litter sizes, but a significant portion of the offspring have low birthweight and abnormal brain development. Evidence from a genome-wide association study of over 150,000 human pregnancies validates the finding because women whose NKG2A receptor is genetically determined to engage their own MHC class I molecules are exposed to lower risk of developing pre-eclampsia, suggesting that maternal uNK cell education is a pre-requisite for a healthy pregnancy and, likely, for healthy offspring too.


Subject(s)
Killer Cells, Natural , Uterus , Pregnancy , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Female , Humans , Uterus/metabolism , Uterus/immunology , Animals , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Receptors, KIR/genetics , Receptors, KIR/metabolism , Immunogenetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily C/genetics , Pre-Eclampsia/immunology , Pre-Eclampsia/genetics
15.
Placenta ; 155: 32-41, 2024 09 26.
Article in English | MEDLINE | ID: mdl-39121585

ABSTRACT

INTRODUCTION: Preeclampsia is a pregnancy-specific disorder characterized by de novo development of hypertension and proteinuria over 20 weeks gestation that has been associated with the dysfunction of trophoblasts. Current evidence suggests that syncytin-1 plays an important role in the non-fusogenic biological activity of trophoblasts, except for specific fusogenic function. However, the underlying mechanism remains unclear. METHODS: The expression and location of syncytin-1 in normal and the late-onset preeclampsia placentas were detected by quantitative real-time PCR, western blotting and immunofluorescence. Morphological and apoptosis analysis were processed in placentas. The ex vivo extravillous explant culture model was used to explore the effect of syncytin-1 on EVT outgrowths. Real-time quantitative PCR and immunoblotting were used to calculate syncytin-1 levels in the trophoblast cells before and after syncytin-1 knockdown or overexpression. CCK-8 assay was used to detect the cell viability. TUNEL staining and immunoblotting were processed in trophoblast cells. Transwell assays and wound healing assays were utilize to assess the invasion and migration of trophoblastic cells. Conditional knockout of syncytin-a mouse model was conducted to present the change of placentas in vivo. The ex vivo extravillous explant culture model was used to explore the effect of syncytin-1 on EVT outgrowths. Western blotting was used to identify the key proteins of PI3K/Akt pathways and invasion-related proteins in trophoblast cells. RESULTS AND DISCUSSION: Here, reduced syncytin-1 was identified in the late-onset preeclampsia placentas. Reduced syncytin-1 may attenuates the EMT process by promoting apoptosis, inhibiting proliferation and invasion by suppressed PI3K/Akt pathway in trophoblast cells. Our findings provide novel insights into the non-fusogenic biological function of reduced syncytin-1 that may be involves in the pathogenesis of preeclampsia.


Subject(s)
Apoptosis , Gene Products, env , Pre-Eclampsia , Pregnancy Proteins , Trophoblasts , Female , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pre-Eclampsia/genetics , Pregnancy , Trophoblasts/metabolism , Trophoblasts/pathology , Apoptosis/physiology , Pregnancy Proteins/metabolism , Pregnancy Proteins/genetics , Humans , Animals , Gene Products, env/metabolism , Gene Products, env/genetics , Mice , Placenta/metabolism , Placenta/pathology , Adult , Mice, Knockout , Cell Movement/physiology , Signal Transduction/physiology
16.
Placenta ; 155: 22-31, 2024 09 26.
Article in English | MEDLINE | ID: mdl-39121584

ABSTRACT

INTRODUCTION: Preeclampsia (PE) is a serious pregnancy-related complication caused by high blood pressure in pregnant women. The severe form has more devastating effects. According to the growing evidence, the placenta is a crucial component in the pathogenesis of PE, and eliminating it will alleviate symptoms. METHODS: GEO's severe preeclampsia placenta microarray datasets; GSE147776, GSE66273, GSE102897, and GSE10588, were chosen to identify differentially expressed genes (DEGs) in different biological pathways. The analysis of hub genes and related non-coding RNAs was done as well. RESULTS: A total of 347 DEGs with adj p-value <0.05 and ǀlog2FoldChangeǀ> 0.5 were discovered between severe PEs and healthy pregnancies, including 204 over-expressed genes and 143 under-expressed genes. The MCC method identified ISG15, IFI44L, MX2, OAS2, MX1, FN1, LDHA, ITGB3, TKT, HK2 genes as the top ten hub genes. Interactions between hub genes and noncoding RNAs were also conducted. The most enriched pathways were as follows; HIF-1 signaling pathway; Pathways in cancer; Alanine, aspartate and glutamate metabolism; Arginine biosynthesis; Human papillomavirus infection; Glycolysis/Gluconeogenesis; Central carbon metabolism in cancer; Valine, leucine and isoleucine degradation; Cysteine and methionine metabolism; and Galactose metabolism. DISCUSSION: This is a secondary data analysis conducted on severe preeclampsia placenta to identify differentially expressed genes, biological pathways, hub-genes, and related noncoding RNAs. Functional studies are crucial to understanding the precise role of these genes in the pathogenesis of PE. Also, accepting a gene as a diagnostic or prognostic marker for early diagnosis and management of PE requires multiple lines of evidence.


Subject(s)
Gene Expression Profiling , Placenta , Pre-Eclampsia , RNA, Untranslated , Humans , Female , Pregnancy , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Placenta/metabolism , RNA, Untranslated/genetics , Signal Transduction/genetics , Microarray Analysis , Gene Regulatory Networks , Transcriptome
17.
Int Immunopharmacol ; 140: 112859, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39121610

ABSTRACT

OBJECTIVE: Caspase-6 is an important regulatory factor in innate immunity, inflammasome activation, and host defense, but its role in preeclampsia (PE) is unknown. This study aims to investigate the mechanism of Caspase-6 in the interaction between PE rats and macrophage-trophoblast cells, in order to provide a new theoretical basis for the treatment of PE. METHODS: Co-cultures of THP-1 cells and HTR8/SVneo cells were employed to investigate the HMGB1 signaling in macrophages (transfection with si-Caspase-6) and HTR8/SVneo cells. The PE rat model was constructed by using the reduced uterine perfusion pressure (RUPP) surgery to explore the therapeutic effects of bone marrow-derived macrophages (BMDM) transfected with si-Caspase-6 in PE rats. ELISA, Western blot, immunofluorescence, etc., were employed to characterize the expression of ferroptosis-related markers. RESULTS: Caspase-6 expression was significantly increased in CD14+ macrophages in the placental tissue of PE rats. Overexpression of Caspase-6 in THP-1 cells induced ferroptosis of HTR8/SVneo cells, but this process was blocked by anti-HMGB1 neutralizing antibody. Knockdown of Caspase-6 in macrophages could alleviate ferroptosis of HTR8/SVneo cells and restore its basic characteristics. Knockdown of Caspase-6 in BMDM downregulated ferroptosis in placental tissue of PE rats through HMGB1, thereby improving the disease phenotype in rats. CONCLUSION: Knocking down Caspase-6 in BMDM regulated the crosstalk between macrophages and HTR8/SVneo cells through HMGB1, inhibiting HTR8/SVneo cell ferroptosis, thereby improving adverse pregnancy outcomes of PE.


Subject(s)
Caspase 6 , Ferroptosis , HMGB1 Protein , Macrophages , Pre-Eclampsia , Trophoblasts , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/immunology , Female , Animals , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Pregnancy , Humans , Macrophages/immunology , Macrophages/metabolism , Rats , Caspase 6/metabolism , Caspase 6/genetics , Trophoblasts/metabolism , THP-1 Cells , Rats, Sprague-Dawley , Disease Models, Animal , Coculture Techniques , Cell Line , Gene Knockdown Techniques
18.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39201703

ABSTRACT

Preeclampsia is a complex pregnancy-related hypertensive disorder which poses significant risks for both maternal and fetal health. Preeclampsia affects 5-8% of pregnancies in the United States, causing a significant public health and economic burden. Despite extensive research, the etiology and pathogenesis of preeclampsia remain elusive, but have been correlated with maternal conditions such as obesity. In recent decades, the incidence of preeclampsia increased along with the prevalence of obesity among women of reproductive age. Maternal obesity has been shown to negatively affect pregnancy in almost all aspects. However, the precise mechanisms by which obesity influences preeclampsia are unclear. Ankyrin repeat and SOCS Box Containing protein 4 (ASB4) is an E3 ubiquitin ligase that can promote the degradation of a wide range of target proteins. ASB4-null mice display a full spectrum of preeclampsia-like phenotypes during pregnancy including hypertension, proteinuria, and decreased litter size. Furthermore, maternal obesity induced by a high-fat diet aggravates preeclampsia-like phenotypes in pregnant mice lacking ASB4. Variants in the ASB4 gene have been associated with obesity in humans, and a functional connection between the ASB4 gene and obesity has been established in mice. This review discusses the connections between preeclampsia, obesity, and ASB4.


Subject(s)
Obesity , Pre-Eclampsia , Suppressor of Cytokine Signaling Proteins , Animals , Female , Humans , Mice , Pregnancy , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Pre-Eclampsia/etiology , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
19.
Pregnancy Hypertens ; 37: 101144, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39111192

ABSTRACT

Preeclampsia or high blood pressure in pregnancy is one of the special disorders during pregnancy. It seems that oxidative stress plays an important role in the occurrence of this disease. The purpose of this study is to investigate the relationship between the A313G polymorphism in exon five of the glutathione S-transferase gene (GSTP1) and the risk of preeclampsia in a case-control study. In this study, blood samples were collected from 70 healthy pregnant women and 70 women with preeclampsia. After genomic DNA extraction, the PCR-RFLP method was performed to check the genotype in GSTP1-A313G and the genotypic frequencies of AA, AG, and GG were determined in all samples. Also, using bioinformatics software, the effect of the above polymorphism on the protein structure was investigated. Statistical analysis for A313G polymorphism showed that AG (OR: 1.1684, 95 % CI: 0.5877-2.3228, p = 0.657) and GG (OR: 1.3793, 95 % CI: 0.3376-5.6359, p = 0.654) genotypes were not associated with risk of preeclampsia in the population of northern Iran. However, bioinformatic analyzes have shown that this polymorphism does have a destructive effect on the protein structure. However, more studies with larger sample sizes are needed to draw firm conclusions.


Subject(s)
Genetic Predisposition to Disease , Glutathione S-Transferase pi , Polymorphism, Single Nucleotide , Pre-Eclampsia , Humans , Female , Pre-Eclampsia/genetics , Pregnancy , Iran/epidemiology , Glutathione S-Transferase pi/genetics , Case-Control Studies , Adult , Risk Factors , Gene Frequency , Young Adult
20.
Medicina (Kaunas) ; 60(8)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39202588

ABSTRACT

Background and objectives: Pre-eclampsia (PE) is a pregnancy-specific condition characterized by significant health risks for pregnant women worldwide due to its status as a multi-organ disorder. High blood pressure (hypertension) with or without proteinuria is usually considered an initial clinical sign of PE. The pathogenesis of pre-eclampsia is highly complex and likely involves multiple factors, including poorly developed uterine spiral arterioles, immunological issues, placental ischemia or infarction, and genetic abnormalities. Inflammatory cytokine production, regulated by cytokine gene polymorphisms, is one of the factors likely contributing to the development of PE. The present study aimed to assess IL-6, IL-1ß, and Apo B-100 gene polymorphism and to evaluate the association of these polymorphisms with PE. Materials and Methods: This cross-sectional observational study involved 99 participants aged 16 to 45 years from Bahawal Victoria Hospital Bahawalpur, Punjab, Pakistan. The participants were divided into three groups: Group 1 (PE with severe hypertension), Group 2 (PE with hypertension), and Group 3 (control), each comprising 33 individuals. Maternal blood samples were collected, DNA was extracted, and molecular genetic analysis of the IL-6, IL-1ß, and Apo B-100 genes was performed using the PCR-RFLP method. Allelic frequencies were compared, and statistical analysis was conducted using SPSS 25, applying the Hardy-Weinberg equation and chi-square test to evaluate the results. Results: There are differences in the distribution of allelic frequencies for IL-6 -174G/C (CC, GC, GG), IL-1ß-511C/T (CC, CT, TT), and Apo B-100 2488 C/T (CC, CT, TT) between pre-eclamptic patients and the control group. The analysis using the Hardy-Weinberg equilibrium and chi-square test showed an association between the IL-6-174 G/C polymorphism and the severity of pre-eclampsia. Conclusions: The polymorphisms of the IL-6, IL-1ß, and Apo B-100 genes revealed different alleles. The IL-6 gene alone was found to be in disequilibrium according to the Hardy-Weinberg equation, indicating a potential link to the severity of pre-eclampsia in the population studied.


Subject(s)
Apolipoprotein B-100 , Interleukin-1beta , Interleukin-6 , Pre-Eclampsia , Humans , Female , Pre-Eclampsia/genetics , Pre-Eclampsia/blood , Pregnancy , Adult , Interleukin-1beta/genetics , Interleukin-1beta/blood , Cross-Sectional Studies , Interleukin-6/genetics , Interleukin-6/blood , Apolipoprotein B-100/genetics , Apolipoprotein B-100/blood , Adolescent , Polymorphism, Genetic , Middle Aged , Pakistan , Young Adult , Genetic Predisposition to Disease
SELECTION OF CITATIONS
SEARCH DETAIL