Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.539
Filter
1.
Int J Mol Sci ; 25(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38674031

ABSTRACT

Hemangioblasts give rise to endothelial progenitor cells (EPCs), which also express the cell surface markers CD133 and c-kit. They may differentiate into the outgrowth endothelial cells (OECs) that control neovascularization in the developing embryo. According to numerous studies, reduced levels of EPCs in circulation have been linked to human cardiovascular disorders. Furthermore, preeclampsia and senescence have been linked to levels of EPCs produced from cord blood. Uncertainties surround how preeclampsia affects the way EPCs function. It is reasonable to speculate that preeclampsia may have an impact on the function of fetal EPCs during the in utero period; however, the present literature suggests that maternal vasculopathies, including preeclampsia, damage fetal circulation. Additionally, the differentiation potential and general activity of EPCs may serve as an indicator of the health of the fetal vascular system as they promote neovascularization and repair during pregnancy. Thus, the purpose of this review is to compare-through the assessment of their quantity, differentiation potency, angiogenic activity, and senescence-the angiogenic function of fetal EPCs obtained from cord blood for normal and pregnancy problems (preeclampsia, gestational diabetes mellitus, and fetal growth restriction). This will shed light on the relationship between the angiogenic function of fetal EPCs and pregnancy complications, which could have an effect on the management of long-term health issues like metabolic and cardiovascular disorders in offspring with abnormal vasculature development.


Subject(s)
Diabetes, Gestational , Endothelial Progenitor Cells , Fetal Blood , Fetal Growth Retardation , Pre-Eclampsia , Humans , Pregnancy , Female , Diabetes, Gestational/metabolism , Diabetes, Gestational/blood , Pre-Eclampsia/blood , Endothelial Progenitor Cells/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Fetal Growth Retardation/pathology , Cell Differentiation
2.
Exp Neurol ; 377: 114781, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636773

ABSTRACT

Chronic hypoxia in utero causes intrauterine growth restriction (IUGR) of the fetus. IUGR infants are known to be at higher risk for neurodevelopmental disorders, but the mechanism is unclear. In this study, we analyzed the structure of the cerebral cortex using IUGR model rats generated through a reduced uterine perfusion pressure operation. IUGR rats exhibited thinner cerebral white matter and enlarged lateral ventricles compared with control rats. Expression of neuron cell markers, Satb2, microtubule-associated protein (MAP)-2, α-tubulin, and nestin was reduced in IUGR rats, indicating that neurons were diminished at various developmental stages in IUGR rats, from neural stem cells to mature neurons. However, there was no increase in apoptosis in IUGR rats. Cells positive for Ki67, a marker of cell proliferation, were reduced in neurons and all glial cells of IUGR rats. In primary neuron cultures, axonal elongation was impaired under hypoxic culture conditions mimicking the intrauterine environment of IUGR infants. Thus, in IUGR rats, chronic hypoxia in utero suppresses the proliferation of neurons and glial cells as well as axonal elongation, resulting in cortical thinning and enlarged lateral ventricles. Thrombopoietin (TPO), a platelet growth factor, inhibited the decrease in neuron number and promoted axon elongation in primary neurons under hypoxic conditions. Intraperitoneal administration of TPO to IUGR rats resulted in increases in the number of NeuN-positive cells and the area coverage of Satb2. In conclusion, suppression of neuronal proliferation and axonal outgrowth in IUGR rats resulted in cortical thinning and enlargement of lateral ventricles. TPO administration might be a novel therapeutic strategy for treating brain dysmaturation in IUGR infants.


Subject(s)
Cell Proliferation , Fetal Growth Retardation , Neuronal Outgrowth , Neurons , Neuroprotective Agents , Rats, Sprague-Dawley , Thrombopoietin , Animals , Fetal Growth Retardation/pathology , Rats , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Female , Cell Proliferation/drug effects , Pregnancy , Neuronal Outgrowth/drug effects , Neuroprotective Agents/pharmacology , Cells, Cultured , Animals, Newborn , Cerebral Cortex/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism
3.
Eur J Obstet Gynecol Reprod Biol ; 296: 158-162, 2024 May.
Article in English | MEDLINE | ID: mdl-38442533

ABSTRACT

INTRODUCTION: Missed abortion (MA) is a type of miscarriage with multiple etiological factors that refers to fetal death with a failure of the retained intrauterine product of conception to be discharged spontaneously. Currently fetal death in missed abortion is categorized according to three main causes: Fetal, placental, and maternal factors. The aim of the current study was to contribute and increase knowledge in clinical practice of late first and second trimester MA (Gestational age: week 11 + 0 - week 20 + 6). MATERIAL AND METHODS: This retrospective case series study includes 794 cases of fetuses and matching placentas sent to the Section of Perinatal Pathology, Department of Pathology, Karolinska Hospital between 2003 and 2019 from five different gynecology departments in the Stockholm region, Sweden. RESULTS: The cases were divided into two groups according to gestational length; gestational week 11 + 0-14 + 6 (group A) and 15 + 0-20 + 6 (group B) respectively, and comparisons were made between groups. Fetal growth restriction and placental pathology were more common in late MA, but number of cases with malformation were higher in early MA. Cord pathology was seen in approximately 40 % of the cases and equally distributed in the gestational weeks included. DISCUSSION: Fetal growth restriction and placental pathology were more common in late second trimester MA. This might demonstrate an early placental dysfunction affecting fetal growth and may be associated to maternal comorbidity such as autoimmune disease and cardiovascular disease. It is advisable to investigate maternal factors more closely after late second trimester MA before a future pregnancy. The risk for recurrent MA is believed to be low in cases of significant cord pathology. CONCLUSION: Cord complications were over-represented in missed abortion suggesting a probable etiopathogenetic link to fetal demise in this condition.


Subject(s)
Abortion, Habitual , Abortion, Missed , Pregnancy , Female , Humans , Placenta/pathology , Abortion, Missed/pathology , Fetal Growth Retardation/pathology , Retrospective Studies , Fetus/pathology , Fetal Death/etiology , Autopsy
4.
J Control Release ; 368: 466-480, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38452820

ABSTRACT

Physiological or pathological hypoperfusion of the placenta is one of the main causes of intrauterine growth restriction (IUGR) which poses a significant risk to the health of the fetus and newborn. Tadalafil, a 5-type phosphodiesterase inhibitor, has previously been found to improve the symptoms of IUGR in various clinical studies. Unfortunately, its clinical utility is hindered by its limited water solubility, rapid metabolism, and lack of specific distribution in target tissues rendering tadalafil unable to maintain long-term placental perfusion. In this study, iRGD-modified tadalafil-loaded liposomes (iRGD-lipo@Tad) featuring a size of approximately 480 nm were designed to rectify the shortcomings of tadalafil. The prepared iRGD-lipo@Tad exhibited superior stability, sustained drug release capacity, and low cytotoxicity. The fluorescence study, tissue slice study, and drug biodistribution study together demonstrated the placenta-anchored ability of iRGD-modified liposomes. This was achieved by a dual approach consisting of the iRGD-mediated placenta-targeting effect and special particle size-mediated placenta resident effect. The pharmacokinetic study revealed a significant improvement in the in vivo process of tadalafil encapsulated by the iRGD-modified liposomes. In comparison to the tadalafil solution, the peak plasma concentration of iRGD-lipo@Tad was significantly increased, and the area under the curve was increased by about 7.88 times. In the pharmacodynamic study, iRGD-lipo@Tad achieved a continuous and efficient improvement of placental blood perfusion. This was achieved by decreasing the ratio of plasma soluble fms-like tyrosine kinase to placental growth factor and increasing the levels of cyclic guanosine monophosphate and nitric oxide. Consequently, iRGD-lipo@Tad resulted in a significant increase in embryo weight and a reduction in the miscarriage rate of N-Nitro-L-arginine methyl ester-induced IUGR pregnant mice without detectable toxicity. In summary, the nanotechnology-assisted therapy strategy presented here not only overcomes the limitations of tadalafil in the clinical treatment of IUGR but also offers new avenues to address the treatment of other placenta-originated diseases.


Subject(s)
Liposomes , Placenta , Humans , Female , Pregnancy , Animals , Mice , Liposomes/metabolism , Tadalafil/therapeutic use , Tadalafil/metabolism , Placenta/metabolism , Placenta/pathology , Fetal Growth Retardation/drug therapy , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Tissue Distribution , Placenta Growth Factor/metabolism , Perfusion
5.
Stem Cell Rev Rep ; 20(4): 996-1014, 2024 May.
Article in English | MEDLINE | ID: mdl-38457061

ABSTRACT

The placenta stands out as a unique, transitory, and multifaceted organ, essential to the optimal growth and maturation of the fetus. Functioning as a vital nexus between the maternal and fetal circulatory systems, it oversees the critical exchange of nutrients and waste. This exchange is facilitated by placental cells, known as trophoblasts, which adeptly invade and remodel uterine blood vessels. Deviations in placental development underpin a slew of pregnancy complications, notably fetal growth restriction (FGR), preeclampsia (PE), recurrent spontaneous abortions (RSA), and preterm birth. Central to placental function and development is epigenetic regulation. Despite its importance, the intricate mechanisms by which epigenetics influence the placenta are not entirely elucidated. Recently, the scientific community has turned its focus to parsing out the epigenetic alterations during placental development, such as variations in promoter DNA methylation, genomic imprints, and shifts in non-coding RNA expression. By establishing correlations between epigenetic shifts in the placenta and pregnancy complications, researchers are unearthing invaluable insights into the biology and pathophysiology of these conditions. This review seeks to synthesize the latest findings on placental epigenetic regulation, spotlighting its crucial role in shaping fetal growth trajectories and development. Through this lens, we underscore the overarching significance of the placenta in the larger narrative of gestational health.


Subject(s)
Epigenesis, Genetic , Placenta , Placentation , Pregnancy Outcome , Humans , Pregnancy , Female , Placentation/genetics , Placenta/metabolism , Pregnancy Outcome/genetics , DNA Methylation/genetics , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Animals
6.
Am J Respir Cell Mol Biol ; 70(5): 400-413, 2024 May.
Article in English | MEDLINE | ID: mdl-38301267

ABSTRACT

Newborns with intrauterine growth restriction (IUGR) have a higher likelihood of developing pulmonary arterial hypertension (PAH) in adulthood. Although there is increasing evidence suggesting that pericytes play a role in regulating myofibroblast transdifferentiation and angiogenesis in malignant and cardiovascular diseases, their involvement in the pathogenesis of IUGR-related pulmonary hypertension and the underlying mechanisms remain incompletely understood. To address this issue, a study was conducted using a Sprague-Dawley rat model of IUGR-related pulmonary hypertension. Our investigation revealed increased proliferation and migration of pulmonary microvascular pericytes in IUGR-related pulmonary hypertension, accompanied by weakened endothelial-pericyte interactions. Through whole-transcriptome sequencing, Ddx5 (DEAD-box protein 5) was identified as one of the hub genes in pericytes. DDX5, a member of the RNA helicase family, plays a role in the regulation of ATP-dependent RNA helicase activities and cellular function. MicroRNAs have been implicated in the pathogenesis of PAH, and microRNA-205 (miR-205) regulates cell proliferation, migration, and angiogenesis. The results of dual-luciferase reporter assays confirmed the specific binding of miR-205 to Ddx5. Mechanistically, miR-205 negatively regulates Ddx5, leading to the degradation of ß-catenin by inhibiting the phosphorylation of Gsk3ß at serine 9. In vitro experiments showed the addition of miR-205 effectively ameliorated pericyte dysfunction. Furthermore, in vivo experiments demonstrated that miR-205 agomir could ameliorate pulmonary hypertension. Our findings indicated that the downregulation of miR-205 expression mediates pericyte dysfunction through the activation of Ddx5. Therefore, targeting the miR-205/Ddx5/p-Gsk3ß/ß-catenin axis could be a promising therapeutic approach for IUGR-related pulmonary hypertension.


Subject(s)
Cell Proliferation , DEAD-box RNA Helicases , Epigenesis, Genetic , Fetal Growth Retardation , Glycogen Synthase Kinase 3 beta , Hypertension, Pulmonary , MicroRNAs , Pericytes , Rats, Sprague-Dawley , Animals , Female , Humans , Male , Rats , beta Catenin/metabolism , beta Catenin/genetics , Cell Movement/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Pericytes/metabolism , Pericytes/pathology
7.
Genes (Basel) ; 15(2)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38397171

ABSTRACT

Bi-allelic pathogenic variations within POLR3A have been associated with a spectrum of hereditary disorders. Among these, a less frequently observed condition is Wiedemann-Rautenstrauch syndrome (WRS), also known as neonatal progeroid syndrome. This syndrome typically manifests neonatally and is characterized by growth retardation, evident generalized lipodystrophy with distinctively localized fat accumulations, sparse scalp hair, and atypical facial features. Our objective was to elucidate the underlying molecular mechanisms of Wiedemann-Rautenstrauch syndrome (WRS). In this study, we present a clinical case of a 7-year-old female patient diagnosed with WRS. Utilizing whole-exome sequencing (WES), we identified a novel missense variant c.3677T>C (p.Leu1226Pro) in the POLR3A gene (NM_007055.4) alongside two cis intronic variants c.1909+22G>A and c.3337-11T>C. Via the analysis of mRNA derived from fibroblasts, we reconfirmed the splicing-affecting nature of the c.3337-11T>C variant. Furthermore, our investigation led to the reclassification of the c.3677T>C (p.Leu1226Pro) variant as a likely pathogenic variant. Therefore, this is the first case demonstrating the molecular genetics of a patient with Wiedemann-Rautenstrauch syndrome from the Russian Federation. A limited number of clinical cases have been documented until this moment; therefore, broadening the linkage between phenotype and molecular changes in the POLR3A gene will significantly contribute to the comprehensive understanding of the molecular basis of POLR3A-related disorders.


Subject(s)
Progeria , Infant, Newborn , Female , Humans , Child , Progeria/genetics , Progeria/diagnosis , Progeria/pathology , Fetal Growth Retardation/pathology , Mutation , Russia , RNA Polymerase III/genetics
8.
Fetal Pediatr Pathol ; 43(3): 198-207, 2024.
Article in English | MEDLINE | ID: mdl-38186330

ABSTRACT

Background: Accurate identification of fetal growth restriction in fetal autopsy is critical for assessing causes of death. We examined the impact of using a chart derived from ultrasound measurements of healthy fetuses (World Health Organization fetal growth chart) versus a chart commonly used by pathologists (Archie et al.) derived from fetal autopsy-based populations in diagnosing small-for-gestational-age (SGA) birth in perinatal deaths. Study Design: We examined perinatal deaths that underwent autopsy at BC Women's Hospital, 2015-2021. Weight centiles were assigned using the ultrasound-based fetal growth chart for birthweight and autopsy-based growth chart for autopsy weight. Results: Among 352 fetuses, 30% were SGA based on the ultrasound-based fetal growth chart versus 17% using the autopsy-based growth chart (p < 0.001). Weight centiles were lower when using the ultrasound-based versus autopsy-based growth chart (median difference of 9 centiles [IQR 2, 20]). Conclusions: Autopsy-based growth charts may under-classify SGA status compared to ultrasound-based fetal growth charts.


Subject(s)
Autopsy , Fetal Growth Retardation , Growth Charts , Infant, Small for Gestational Age , Humans , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/pathology , Autopsy/methods , Female , Infant, Newborn , Pregnancy , Ultrasonography, Prenatal/methods , Fetal Development/physiology , Gestational Age , Birth Weight
9.
Placenta ; 147: 21-27, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38278001

ABSTRACT

INTRODUCTION: Gestational diabetes mellitus (GDM) exerts a great impact on the placenta and reflects changes on placentas both morphological and functionally. The aims of this study are to evaluate the prevalence of placental histopathological lesions in pregnancies complicated by GDM compared to gestational age-matched controls, and their association with maternal and fetal complications. METHODS: Fifty-four singleton GDM-complicated pregnancies were recruited and compared to 33 consecutive normal pregnancies. Two pathologists, blinded to all clinical data, reviewed and evaluated all histological samples of the placentas in accordance with Amsterdam criteria. Relevant demographic, clinical data and primary birth outcomes were recorded. RESULTS: A myriad of histomorphological abnormalities, including chronic inflammation (n = 9/54, p = 0.031), histological chorioamnionitis (n = 23/54, p < 0.001), umbilical/chorionic vasculitis (n = 9/54, p = 0.031), changes related to maternal vascular malperfusion (n = 22/54, p = 0.003), chorangiosis (n = 10/54, p = 0.046) and villous dysmaturity (n = 9/54, p = 0.012) were observed more frequently in the GDM placentas compared to the controls. Additionally, GDM significantly increased the risk of fetal complications, including macrosomia/fetal growth restriction (n = 13/54, p = 0.004). DISCUSSION: Histoarchitectural abnormalities were observed more frequently in placentas of GDM pregnancies compared to the controls. Our findings support the hypothesis that diabetic-induced damage in the placental function may be associated with the increased in fetal growth disorders in GDM-complicated pregnancies.


Subject(s)
Diabetes, Gestational , Placenta , Pregnancy , Female , Humans , Placenta/pathology , Diabetes, Gestational/pathology , Fetal Macrosomia , Fetal Growth Retardation/pathology
10.
Arch Gynecol Obstet ; 309(4): 1305-1313, 2024 Apr.
Article in English | MEDLINE | ID: mdl-36933038

ABSTRACT

PURPOSE: Although small for gestational age (SGA) does not cause adverse perinatal outcomes, the placental pathology for fetal growth restricted (FGR) and SGA fetuses is still unknown. The aim of this study is to evaluate the differences between placentas of early onset FGR, late onset FGR, SGA, and appropriate for gestational age (AGA) pregnancies in the manner of microvasculature and expression of anti-angiogenic PEDF factor and CD68. METHODS: The study included four groups (early onset FGR, late onset FGR, SGA and AGA). Placental samples were obtained just after labor in all of the groups. Degenerative criteria were investigated with Hematoxylin-eosin staining. Immunohistochemical evaluation with H score and m RNA levels of Cluster of differentiation 68 (CD68) and pigment epithelium derived factor (PEDF) were performed for each group. RESULTS: The highest levels of degeneration were detected in the early onset FGR group. In means of degeneration SGA placentas were found to be worse than the AGA placentas. The intensity of PEDF and CD68 were significant in early FGR, the late FGR and SGA groups compared to the AGA group (p < 0.001). The mRNA level results of the PEDF and CD68 were also parallel to the immunostaining results. CONCLUSION: Although SGA fetuses are considered constitutionally small, the SGA placentas also demonstrated signs of degeneration similar to the FGR placentas. These degenerative signs were not seen among the AGA placentas.


Subject(s)
Infant, Newborn, Diseases , Placenta , Infant, Newborn , Pregnancy , Female , Humans , Placenta/pathology , Gestational Age , Fetal Growth Retardation/pathology , Infant, Small for Gestational Age , Infant, Newborn, Diseases/pathology , Parturition , Fetus
11.
Placenta ; 145: 60-64, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38071790

ABSTRACT

Placental histopathologic lesions are dichotomized into "present" or "absent" and have limited inter-rater reliability. Continuous metrics are needed to characterize placental health and function. Tissue sections (N = 64) of human placenta were stained with CD34 antibody and hematoxylin. Proportion of the villous space occupied by fetal vascular endothelium (%FVE; pixels positive for CD34/total pixels) was evaluated for effect sizes associated with pregnancy outcomes, smoking status, and subtypes of lesions (n = 30). Time to fixation>60 min significantly increased the quantification. Large effect sizes were found between %FVE and both preterm birth and intrauterine growth restriction. These results demonstrate proof-of-concept for this vascular estimation.


Subject(s)
Placenta Diseases , Premature Birth , Infant, Newborn , Pregnancy , Female , Humans , Placenta/pathology , Reproducibility of Results , Premature Birth/pathology , Pregnancy Outcome , Placenta Diseases/diagnosis , Placenta Diseases/pathology , Fetal Growth Retardation/pathology
12.
Proteomics ; 24(7): e2300260, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38059784

ABSTRACT

Intrauterine growth restriction (IUGR) is associated with increased risk of cardiometabolic disease later in life and has been shown to affect female and male offspring differently, but the mechanisms remain unclear. The purpose of this study was to identify proteomic differences and metabolic risk markers in IUGR male and female neonates when compared to appropriate for gestational age (AGA) babies that will provide a better understanding of IUGR pathogenesis and its associated risks. Our results revealed alterations in IUGR cord plasma proteomes with most of the differentially abundant proteins implicated in peroxisome pathways. This effect was evident in females but not in males. Furthermore, we observed that catalase activity, a peroxisomal enzyme, was significantly increased in females (p < 0.05) but unchanged in males. Finally, we identified risk proteins associated with obesity, type-2 diabetes, and glucose intolerance such as EGF containing fibulin extracellular matrix protein 1 (EFEMP1), proprotein convertase subtilisin/kexin type 9 (PCSK9) and transforming growth factor beta receptor 3 (TGFBR3) proteins unique to females while coagulation factor IX (C9) and retinol binding protein 4 (RBP4) are unique in males. In conclusion, IUGR may display sexual dimorphism which may be associated with differences in lifelong risk for cardiometabolic disease between males and females.


Subject(s)
Cardiovascular Diseases , Fetal Growth Retardation , Infant, Newborn , Infant , Humans , Male , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Proprotein Convertase 9/metabolism , Proteomics , Retinol-Binding Proteins, Plasma , Extracellular Matrix Proteins/metabolism
13.
Reprod Sci ; 31(4): 997-1005, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37973775

ABSTRACT

Tumor necrosis factor-α (TNF-α) antagonists are highly effective in controlling autoimmune diseases. This has led to speculation that they might also be useful in treating inflammatory placental conditions, such as chronic villitis of unknown etiology (VUE). VUE affects 10-15% of term placentas and is associated with recurrent fetal growth restriction (FGR) and pregnancy loss. We aimed to evaluate outcomes in patients with autoimmune diseases with and without anti-TNF-α biologic exposure during gestation. This retrospective cohort study compared pregnant women with autoimmune disease taking anti-TNF-α biologics (n = 89) to pregnant women with autoimmune disease but not taking a biologic (n = 53). We extracted data on all patients meeting our inclusion criteria over a 20-year period. Our primary outcome was the diagnosis of VUE by histology. Our secondary outcomes were maternal and neonatal complications such as preeclampsia, FGR, and neonatal intensive care admission. Kruskal-Wallis and chi-squared tests were performed as appropriate for statistical analysis. Maternal characteristics were comparable between groups, and there was no increase in adverse pregnancy outcomes based on anti-TNF-α treatment. Exposure to anti-TNF-α therapy had no significant effect on the incidence of VUE or other obstetric complications. Within the cohort exposed to anti-TNF-α biologics during pregnancy, the rate of VUE was 9.3%, which is comparable to the reported general population risk. Our data support the safety profile of biologic use in pregnancy.


Subject(s)
Autoimmune Diseases , Biological Products , Chorioamnionitis , Placenta Diseases , Infant, Newborn , Humans , Pregnancy , Female , Placenta/pathology , Tumor Necrosis Factor Inhibitors/adverse effects , Placenta Diseases/diagnosis , Chorionic Villi/pathology , Retrospective Studies , Pregnancy Outcome , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/pathology , Autoimmune Diseases/diagnosis , Autoimmune Diseases/drug therapy , Autoimmune Diseases/complications , Biological Products/adverse effects
14.
Pediatr Dev Pathol ; 27(1): 52-58, 2024.
Article in English | MEDLINE | ID: mdl-37771135

ABSTRACT

PURPOSE AND CONTEXT: Umbilical cord abnormalities with clinical signs of cord compromise are frequently associated with fetal vascular malperfusion (FVM). Single umbilical artery (SUA) has been reported to be associated with high-grade FVM in fetal growth restriction but not in an unselected population; our study aimed to address this issue. METHODS: Clinical and placental phenotypes of 55 consecutive placentas with SUA (Group 1) were compared with those of 655 placentas with 3-vessel umbilical cord (Group 2) from patients who were in the second half of their pregnancy. The placentas were histologically examined using hematoxylin and eosin (H&E) staining and CD 34 immunostaining. KEY RESULTS: Several umbilical cord phenotypes and high-grade distal FVM, based on H&E staining and endothelial fragmentation by CD34 were significantly more common in Group 1, whereas decidual clusters of multinucleate trophoblasts were more common in Group 2. Notably, H&E staining or CD34 immunostaining evaluated separately showed that high-grade distal FVM was more common in Group 1 than in Group 2, but the difference was not statistically significant. CONCLUSIONS: SUA predisposes to remote, advanced, and recent high-grade distal villous FVM, with a pathogenesis partly different from that of stasis-induced FVM, likely related to fetal anomalies associated with SUA.


Subject(s)
Placenta Diseases , Single Umbilical Artery , Pregnancy , Humans , Female , Placenta/pathology , Single Umbilical Artery/pathology , Placenta Diseases/pathology , Umbilical Cord/pathology , Fetal Growth Retardation/pathology , Antigens, CD34
15.
Placenta ; 145: 19-26, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38011757

ABSTRACT

INTRODUCTION: Hypertensive disorders of pregnancy (HDP) and fetal growth restriction (FGR) are common obstetrical complications, often with pathological features of maternal vascular malperfusion (MVM) in the placenta. Currently, clinical placental pathology methods involve a manual visual examination of histology sections, a practice that can be resource-intensive and demonstrates moderate-to-poor inter-pathologist agreement on diagnostic outcomes, dependant on the degree of pathologist sub-specialty training. METHODS: This study aims to apply machine learning (ML) feature extraction methods to classify digital images of placental histopathology specimens, collected from cases of HDP [pregnancy induced hypertension (PIH), preeclampsia (PE), PE + FGR], normotensive FGR, and healthy pregnancies, according to the presence or absence of MVM lesions. 159 digital images were captured from histological placental specimens, manually scored for MVM lesions (MVM- or MVM+) and used to develop a support vector machine (SVM) classifier model, using features extracted from pre-trained ResNet18. The model was trained with data augmentation and shuffling, with the performance assessed for patch-level and image-level classification through measurements of accuracy, precision, and recall using confusion matrices. RESULTS: The SVM model demonstrated accuracies of 70 % and 79 % for patch-level and image-level MVM classification, respectively, with poorest performance observed on images with borderline MVM presence, as determined through post hoc observation. DISCUSSION: The results are promising for the integration of ML methods into the placental histopathological examination process. Using this study as a proof-of-concept will lead our group and others to carry ML models further in placental histopathology.


Subject(s)
Hypertension, Pregnancy-Induced , Pre-Eclampsia , Pregnancy , Female , Humans , Placenta/pathology , Pregnancy Outcome , Retrospective Studies , Pre-Eclampsia/pathology , Hypertension, Pregnancy-Induced/pathology , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/pathology
16.
Pediatr Dev Pathol ; 27(1): 23-31, 2024.
Article in English | MEDLINE | ID: mdl-37818682

ABSTRACT

INTRODUCTION: Skeletal growth arrest lines (GAL) are transverse lines of metaphyseal radiodensity accompanying episodic severe physiological stress. They are poorly described in fetal remains. MATERIALS AND METHODS: We searched our autopsy practice for instances of fetal GAL in post mortem radiology, and correlated them with long bone histology and placental pathology. We describe the appearance, distribution, and pathology of GAL in a cohort of fetal autopsies, and compare the placental pathology accompanying GAL to the placental pathology of asymmetrical growth restriction (AGR) in the same time period. RESULTS: In 2108 consecutive fetal post mortems, we found 20 cases with GAL. About 16 were in singletons with AGR. In these 16, the distribution of placental pathologies was similar to a contemporaneous cohort of 113 cases with AGR. Of the remaining 4, two twins out of 9 sets of monochorionic twins with AGR demonstrated GAL. One case of GAL had symmetrical growth restriction with cytomegalovirus infection, and one case had no AGR and an old, unexplained retroplacental hemorrhage. On histology, GAL are characterized by a region of mineralized chondroid, which is variably incorporated into irregular trabecular bone. DISCUSSION: GALs accompany a variety of placental pathologies and twin-twin transfusion, suggesting episodic disease progression.


Subject(s)
Fetofetal Transfusion , Placenta , Pregnancy , Female , Humans , Placenta/pathology , Fetofetal Transfusion/pathology , Fetus/pathology , Fetal Growth Retardation/pathology
17.
Placenta ; 146: 30-41, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38160601

ABSTRACT

INTRODUCTION: Fetal growth restriction (FGR) can lead to fetal mental development abnormalities, malformations, and even intrauterine death. Defects in the trophoblasts at the maternal-fetal interface may contribute to FGR. However, the impact of trophoblasts on FGR is still not well understood. Therefore, the objective of this study is to characterize the heterogeneity of placental cells at the single-cell level and investigate the role of trophoblast subtypes in the pathogenesis of FGR at the cellular and molecular levels. METHODS: Single-cell RNA sequencing was performed on the maternal side of placentas from two normal pregnant women and two pregnant women with FGR. Lentivirus transfection was used to establish a FN1 knockout model in trophoblast HTR-8-Svneo cells. The effect of FN1 knockout on cell migration and invasion of HTR-8-Svneo cells was assessed through wound healing and transwell assays. RESULTS: Nine cell types were annotated in 39,161 cells derived from single-cell RNA sequencing. The FGR group exhibited a decrease in the percentage of trophoblasts, especially in subtype of extravillous trophoblasts (EVTs). The expression of FN1 was reduced in trophoblasts and EVTs. Furthermore, the protein expression levels of FN1 in the placentas of FGR patients were significantly lower than those of normal pregnant women. The cell migration and invasion ability of HTR-8-Svneo cells were inhibited after the knockdown of FN1. DISCUSSION: The dysregulation of the trophoblast subtype-EVTs is involved in placental dysplasia related to FGR. The association between aberrant placental trophoblasts and reduced FN1 expression may contribute to insufficient remodeling of spiral arteries and the formation of FGR.


Subject(s)
Fetal Growth Retardation , Placenta , Female , Humans , Pregnancy , Cell Line , Cell Movement , Fetal Growth Retardation/pathology , Placenta/metabolism , Sequence Analysis, RNA , Trophoblasts/metabolism
18.
Environ Health Perspect ; 131(11): 117011, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37995155

ABSTRACT

BACKGROUND: Perfluorohexane sulfonate (PFHxS) is a frequently detected per- and polyfluoroalkyl substance in most populations, including in individuals who are pregnant, a period critical for early life development. Despite epidemiological evidence of exposure, developmental toxicity, particularly at realistic human exposures, remains understudied. OBJECTIVES: We evaluated the effect of gestational exposure to human-relevant body burden of PFHxS on fetal and placental development and explored mechanisms of action combining alternative splicing (AS) and gene expression (GE) analyses. METHODS: Pregnant ICR mice were exposed to 0, 0.03, and 0.3µg/kg/day from gestational day 7 to day 17 via oral gavage. Upon euthanasia, PFHxS distribution was measured using liquid chromatography-tandem mass spectrometry. Maternal and fetal phenotypes were recorded, and histopathology was examined for placenta impairment. Multiomics was adopted by combining AS and GE analyses to unveil disruptions in mRNA quality and quantity. The key metabolite transporters were validated by quantitative real-time PCR (qRT-PCR) for quantification and three-dimensional (3D) structural simulation by AlphaFold2. Targeted metabolomics based on liquid chromatography-tandem mass spectrometry was used to detect amino acid and amides levels in the placenta. RESULTS: Pups developmentally exposed to PFHxS exhibited signs of intrauterine growth restriction (IUGR), characterized by smaller fetal weight and body length (p<0.01) compared to control mice. PFHxS concentration in maternal plasma was 5.01±0.54 ng/mL. PFHxS trans-placenta distribution suggested dose-dependent transfer through placental barrier. Histopathology of placenta of exposed dams showed placental dysplasia, manifested with an attenuated labyrinthine layer area and deescalated blood sinus counts and placental vascular development index marker CD34. Combined GE and AS analyses pinpointed differences in genes associated with key biological processes of placental development, proliferation, metabolism, and transport in placenta of exposed dams compared to that of control dams. Further detection of placental key transporter gene expression, protein structure simulation, and amino acid and amide metabolites levels suggested that PFHxS exposure during pregnancy led to impairment of placental amino acid transportation. DISCUSSION: The findings from this study suggest that exposure to human-relevant very-low-dose PFHxS during pregnancy in mice caused IUGR, likely via downregulating of placental amino acid transporters, thereby impairing placental amino acid transportation, resulting in impairment of placental development. Our findings confirm epidemiological findings and call for future attention on the health risk of this persistent yet ubiquitous chemical in the early developmental stage and provide a new approach for understanding gene expression from both quantitative and qualitative omics approaches in toxicological studies. https://doi.org/10.1289/EHP13217.


Subject(s)
Fluorocarbons , Placentation , Humans , Pregnancy , Mice , Animals , Female , Placenta , Alternative Splicing , Mice, Inbred ICR , Fluorocarbons/toxicity , Fluorocarbons/metabolism , Alkanesulfonates/metabolism , Alkanesulfonates/pharmacology , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Amino Acids/metabolism , Amino Acids/pharmacology , Gene Expression Profiling
19.
Eur J Med Genet ; 66(11): 104852, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37758168

ABSTRACT

Defects in L-serine biosynthesis are a group of autosomal recessive diseases resulting in a wide phenotypic spectrum ranging from viable to lethal presentations and caused by variants in the three genes encoding the L-serine biosynthesis enzymes, PHGDH, PSAT1, and PSPH. Neu-Laxova syndrome (NLS) is the fetal form of this group, characterized by multiple congenital anomalies including severe intrauterine growth retardation, cutaneous lesions extending from ichthyosis to severe restrictive dermopathy with ectropion and eclabion, edema, microcephaly, central nervous system abnormalities, and flexion contractures. Here we report on two unrelated fetuses with an attenuated phenotype of NLS, that initially evoked Taybi-Linder syndrome. They carry biallelic pathogenic variants in the PHGDH gene. These observations expand the phenotypic continuum of L-serine biosynthesis defects, and illustrate the phenotypic overlap between NLS and microcephalic primordial dwarfism.


Subject(s)
Dwarfism , Microcephaly , Female , Humans , Microcephaly/genetics , Microcephaly/pathology , Fetus/pathology , Dwarfism/genetics , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Serine
20.
Placenta ; 139: 159-171, 2023 08.
Article in English | MEDLINE | ID: mdl-37406553

ABSTRACT

INTRODUCTION: Fetal growth restriction (FGR) is a common complication of pregnancy. Lipid metabolism and distribution may contribute to the progression of FGR. However, the metabolism-related mechanisms of FGR remain unclear. The aim of this study was to identify metabolic profiles associated with FGR, as well as probable genes and signaling pathways. METHODS: Metabolomic profiles at the maternal-fetal interface (including the placenta, maternal and fetal serum) from pregnant women with (n = 35) and without (n = 35) FGR were analyzed by gas chromatography-mass spectrometry (GC-MS). Combined with differentially expressed genes (DEGs) from the GSE35574 dataset, analysis was performed for differential metabolites, and identified by the Metabo Analyst dataset. Finally, the pathology and screened DEGs were further identified. RESULTS: The results showed that fatty acids (FAs) accumulated in the placenta and decreased in fetal blood in FGR cases compared to controls. The linoleic acid metabolism was the focus of placental differential metabolites and genes enrichment analysis. In this pathway, phosphatidylcholine can interact with PLA2G2A and PLA2G4C, and 12(13)-EpOME can interact with CYP2J2. PLA2G2A and CYP2J2 were elevated, and PLA2G4C was decreased in the FGR placenta. DISCUSSION: In conclusion, accumulation of FAs in the placental ischemic environments, may involve linoleic acid metabolism, which may be regulated by PLA2G2A, CYP2J2, and PLA2G4C. This study may contribute to understanding the underlying metabolic and molecular mechanisms of FGR.


Subject(s)
Fetal Growth Retardation , Placenta , Pregnancy , Female , Humans , Fetal Growth Retardation/pathology , Placenta/metabolism , Lipid Metabolism , Cytochrome P-450 CYP2J2 , Linoleic Acids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...