Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 328
Filtrar
1.
Placenta ; 150: 52-61, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38593636

RESUMEN

INTRODUCTION: Does an elevation in d-Galactose (D-Gal) levels within the body contribute to abnormal embryonic development and placental dysfunction during pregnancy? METHODS: Mouse embryos were cultivated to the blastocyst stage under varying concentrations of D-Gal. The blastocyst formation rate was measured, and the levels of reactive oxygen species (ROS), sirtuin 1 (SIRT1), and forkhead box O3a (FOXO3a) in blastocysts were assessed. Mice were intraperitoneally injected with either saline or D-Gal with or without SRT1720. On the 14th day of pregnancy, the fetal absorption rate and placental weight were recorded. Placental levels of superoxide dismutase (SOD) and malondialdehyde (MDA) were determined. The expression of senescence-related factors, such as senescence-associated ß-galactosidase (SA-ß-gal) in the placenta was examined, and the expression of placental SIRT1, FOXO3a and p21 was evaluated by immunohistochemistry and Western blotting. RESULTS: D-Gal adversely affects early embryonic development in vitro, resulting in a decreased blastocyst formation rate. Furthermore, D-Gal downregulates SIRT1 and FOXO3a while increasing ROS levels in blastocysts. Concurrently, D-Gal induces placental dysfunction, characterized by an elevated fetal absorption rate, reduced placental weight, diminished SOD activity, and increased MDA content. The senescence-related factor SA-ß-gal was detected in the placenta, along with altered expression of placental SIRT1, FOXO3a, and p21. The SIRT1 agonist SRT1720 mitigated this damage by increasing SIRT1 and FOXO3a expression. DISCUSSION: The inhibition of early embryonic development and placental dysfunction induced by D-Gal may be attributed to the dysregulation of SIRT1. Activating SIRT1 emerges as a potentially effective strategy for alleviating the adverse effects of D-Gal exposure.


Asunto(s)
Desarrollo Embrionario , Proteína Forkhead Box O3 , Galactosa , Placenta , Especies Reactivas de Oxígeno , Sirtuina 1 , Animales , Proteína Forkhead Box O3/metabolismo , Femenino , Sirtuina 1/metabolismo , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Ratones , Desarrollo Embrionario/efectos de los fármacos , Placenta/metabolismo , Placenta/efectos de los fármacos , Enfermedades Placentarias/metabolismo , Enfermedades Placentarias/inducido químicamente
2.
Methods Mol Biol ; 2767: 175-188, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-36773273

RESUMEN

Human pluripotent stem cells (hPSCs) form an ideal system to study the formation of placental cells, from an undifferentiated human embryonic stem cell state. The conventional human in vitro model systems to study the human placenta cannot be employed for understanding placental dysfunctions or the development of specialized placental cell types. Hence, human PSCs make an ideal model system to study human placental development and disorders. Here, we describe an efficient and validated protocol to reproducibly study the formation of human cytotrophoblasts (CTBs) and syncytiotrophoblast (STBs) from undifferentiated hPSCs. CTBs are the trophoblast stem cells that can differentiate into specialized placental cell types such as STBs. The multinucleated STB plays vital role in the exchange of nutrients and gases across the placenta and secretes several hormones during pregnancy, such as human chorionic gonadotropin ß (hCGß). Here we describe two methods of seeding the hPSCs: chemical (clumps method) and enzymatic methods (single cells) to differentiate them to CTB and STB, activating BMP (B) signaling and inhibiting ACTIVIN/NODAL and FGF signaling pathways (2i), thus naming our protocol as "B2i" (Sudheer et al., Stem Cells Dev 21:2987-3000, 2012). This protocol forms the perfect model system for understanding in vitro placentation, modeling diseases arising from abnormal placentation that cause complications such as miscarriage, preeclampsia or intrauterine growth restriction (IUGR), and drug discovery for placental disorders.


Asunto(s)
Enfermedades Placentarias , Células Madre Pluripotentes , Humanos , Embarazo , Femenino , Placenta , Trofoblastos , Placentación/fisiología , Diferenciación Celular/fisiología , Enfermedades Placentarias/metabolismo
3.
Placenta ; 143: 80-86, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37864887

RESUMEN

INTRODUCTION: Our understanding of the etiology of preterm birth (PTB) is incomplete; however, recent evidence has found a strong association between placental dysfunction and PTB. Altered placental metabolism may precede placental dysfunction and therefore the study of placental metabolic profiles could identify early biomarkers of PTB. In this study, we evaluated the placental metabolome in PTB in intact tissue samples using nuclear magnetic resonance (NMR) and spectral editing. METHODS: Placental tissue samples were collected from nine term pregnancies and nine preterm pregnancies (<37 weeks' gestation). 1H NMR experiments on unprocessed tissue samples were performed using a high field magnet (500 MHz spectrometer) and a comprehensive multiphase NMR probe. The relative concentrations of 23 metabolites were corrected for gestational age and compared between groups. RESULTS: The relative concentration of valine, glutamate and creatine were significantly decreased while alanine, choline and glucose were elevated in placentas from PTB pregnancies compared to controls (p < 0.05). Multivariate analysis using principal component analysis showed the PTB and control groups were significantly separated (p < 0.0001) and pathway analysis identified perturbations in the glycine, serine and threonine metabolism, aminoacyl-tRNA biosynthesis and valine, leucine and isoleucine biosynthesis pathways. CONCLUSION: PTB is associated with significant alterations in placental metabolism. This study helps improve our understanding of the etiology of PTB. It also highlights the potential for small molecule metabolites to serve as placental metabolic biomarkers to aid in the prediction and diagnosis of PTB. The results can be translated to clinical use via in utero magnetic resonance spectroscopy.


Asunto(s)
Enfermedades Placentarias , Nacimiento Prematuro , Embarazo , Recién Nacido , Femenino , Humanos , Placenta/diagnóstico por imagen , Placenta/metabolismo , Nacimiento Prematuro/metabolismo , Espectroscopía de Resonancia Magnética , Enfermedades Placentarias/metabolismo , Biomarcadores/metabolismo , Valina/metabolismo
4.
Placenta ; 143: 12-15, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37793322

RESUMEN

The placenta remains the key organ to pregnancy complications, such as preeclampsia, contrarily the pathophysiology underlying the placental dysfunctions remains elusive. Here, we present our Disease Map "NaviCenta", which is an online resource based on the interactions between tissues, cellular compartments, and molecules that mediate disease-related processes in the placenta. We built cellular and molecular interaction networks based upon manual curation and annotation of publicly available information in the scientific literature, pathways resources, and Omics data. NaviCenta (Navigate the plaCenta) serves as an open access, spatio-temporal, multi-scale knowledge base, and analytical tool for enhanced interpretation and hypothesis testing on various placental disease phenotypes.


Asunto(s)
Enfermedades Placentarias , Preeclampsia , Complicaciones del Embarazo , Embarazo , Femenino , Humanos , Placenta/metabolismo , Enfermedades Placentarias/metabolismo , Complicaciones del Embarazo/metabolismo , Preeclampsia/metabolismo
5.
Int J Mol Sci ; 24(11)2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37298167

RESUMEN

Eosinophilic/T-cell chorionic vasculitis (ETCV) is an idiopathic lesion composed of eosinophils, CD3+ T lymphocytes, and histiocytes. In twins, ETCV may affect only one chorionic plate, a feature defined as "discordant". We present a case of ETCV discordance in a diamniotic dichorionic placenta at 38 weeks of gestation, in which the female twin was small for gestational age, weighing 2670 g (25th percentile). The corresponding placental territory presented ETCV in two close chorionic vessels with concordance of the fetal inflammatory response. Immunohistochemistry showed an abundance of CD3+/CD4+/CD25+T lymphocytes, CD68 PG M1+ macrophages, and scattered CD8+ T cells with focal TIA-1 positivity. Granzyme B, CD20 B lymphocytes, and CD56 natural killer cells were negative. High-grade villitis of unknown etiology (VUE) was additionally found and displayed comparable ETCV findings, except for an equivalent ratio of CD4+/CD8+ T cells, but TIA-1 was focally expressed. VUE was associated with chronic histiocytic intervillositis (CHI). The combination of ETCV, VUE, and CHI may have been responsible for reduced fetal growth. Concordance was observed in the ETCV and TIA-1 expression, both in ETCV and in VUE, which is a maternal response. These findings may suggest a common antigen or chemokine pathway to which both mother and fetus accordingly responded.


Asunto(s)
Enfermedades Placentarias , Vasculitis , Femenino , Embarazo , Humanos , Placenta/metabolismo , Enfermedades Placentarias/metabolismo , Corion/metabolismo , Linfocitos T CD8-positivos , Vellosidades Coriónicas/metabolismo
6.
Reproduction ; 166(2): R25-R38, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37318094

RESUMEN

In brief: Placental oxidative stress contributes to both normal and abnormal placentation during pregnancy. This review discusses the potential consequence of oxidative stress-induced placental dysfunction on pregnancies complicated by fetal death and pregnancies with a high risk of fetal death. Abstract: The placenta is a source of reactive oxygen free radicals due to the oxidative metabolism required to meet the demands of the growing fetus. The placenta has an array of efficient antioxidant defense systems to deal with rising oxidative stress created by free radicals during pregnancy. Properly controlled physiological (low-level) free radical production is a necessary part of cellular signaling pathways and downstream activities during normal placental development; however, poorly controlled oxidative stress can cause aberrant placentation, immune disturbances and placental dysfunction. Abnormal placental function and immune disturbances are linked to many pregnancy-related disorders, including early and recurrent pregnancy loss, fetal death, spontaneous preterm birth, preeclampsia and fetal growth restriction. This review discusses the role of placental oxidative stress in both normal and pathological settings. Finally, based on previously published work, this review presents multiple lines of evidence for the strong association between oxidative stress and adverse pregnancy outcomes, including fetal death and pregnancies with a high risk of fetal death.


Asunto(s)
Enfermedades Placentarias , Preeclampsia , Complicaciones del Embarazo , Nacimiento Prematuro , Embarazo , Femenino , Recién Nacido , Humanos , Placenta/metabolismo , Nacimiento Prematuro/metabolismo , Placentación , Estrés Oxidativo/fisiología , Complicaciones del Embarazo/metabolismo , Enfermedades Placentarias/metabolismo , Muerte Fetal/etiología , Preeclampsia/metabolismo , Retardo del Crecimiento Fetal/metabolismo
7.
Hypertension ; 80(6): 1363-1374, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36987911

RESUMEN

BACKGROUND: Placental disorders contribute to pregnancy complications, including preeclampsia and fetal growth restriction (FGR), but debate regarding their specific pathobiology persists. Our objective was to apply transcriptomics with weighted gene correlation network analysis to further clarify the placental dysfunction in these conditions. METHODS: We performed RNA sequencing with weighted gene correlation network analysis using human placental samples (n=30), separated into villous tissue and decidua basalis, and clinically grouped as follows: (1) early-onset preeclampsia (EOPE)+FGR (n=7); (2) normotensive, nonanomalous preterm FGR (n=5); (2) EOPE without FGR (n=8); (4) spontaneous idiopathic preterm birth (n=5) matched for gestational age; and (5) uncomplicated term births (n=5). Our data was compared with RNA sequencing data sets from public databases (GSE114691, GSE148241, and PRJEB30656; n=130 samples). RESULTS: We identified 14 correlated gene modules in our specimens, of which most were significantly correlated with birthweight and maternal blood pressure. Of the 3 network modules consistently predictive of EOPE±FGR across data sets, we prioritized a coexpression gene group enriched for hypoxia-response and metabolic pathways for further investigation. Cluster analysis based on transcripts from this module and the glycolysis/gluconeogenesis metabolic pathway consistently distinguished a subset of EOPE±FGR samples with an expression signature suggesting modified tissue bioenergetics. We demonstrated that the expression ratios of LDHA/LDHB and PDK1/GOT1 could be used as surrogate indices for the larger panels of genes in identifying this subgroup. CONCLUSIONS: We provide novel evidence for a molecular subphenotype consistent with a glycolytic metabolic shift that occurs more frequently but not universally in placental specimens of EOPE±FGR.


Asunto(s)
Enfermedades Placentarias , Preeclampsia , Nacimiento Prematuro , Humanos , Embarazo , Recién Nacido , Femenino , Placenta/metabolismo , Retardo del Crecimiento Fetal , Transcriptoma , Preeclampsia/metabolismo , Nacimiento Prematuro/metabolismo , Enfermedades Placentarias/metabolismo
8.
Reprod Sci ; 30(7): 2079-2086, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36930425

RESUMEN

Ferroptosis is a recently identified form of programmed cell death which is different from apoptosis, pyroptosis, necrosis, and autophagy. It is uniquely defined by redox-active iron-dependent hydroxy-peroxidation of polyunsaturated fatty acid (PUFA)-containing phospholipids and a loss of lipid peroxidation repair capacity. Ferroptosis has recently been implicated in multiple human diseases, such as tumors, ischemia-reperfusion injury, acute kidney injury, neurological diseases, and asthma among others. Intriguingly, ferroptosis is associated with placental physiology and trophoblast injury. Circumstances such as accumulation of lipid reactive oxygen species (ROS) due to hypoxia-reperfusion and anoxia-reoxygenation of trophoblast during placental development, the abundance of trophoblastic iron and PUFA, physiological uterine contractions, or pathological placental bed perfusion, cause placental trophoblasts' susceptibility to ferroptosis. Ferroptosis of trophoblast can cause placental dysfunction, which may be involved in the occurrence and development of placenta-related diseases such as gestational diabetes mellitus, preeclampsia, fetal growth restriction, preterm birth, and abortion. The regulatory mechanisms of trophoblastic ferroptosis still need to be explored further. Here, we summarize the latest progress in trophoblastic ferroptosis research on placental-related diseases, provide references for further understanding of its pathogenesis, and propose new strategies for the prevention and treatment of placental-related diseases.


Asunto(s)
Ferroptosis , Enfermedades Placentarias , Nacimiento Prematuro , Recién Nacido , Embarazo , Femenino , Humanos , Placenta/metabolismo , Nacimiento Prematuro/metabolismo , Apoptosis , Peroxidación de Lípido , Hierro , Enfermedades Placentarias/metabolismo , Hipoxia/metabolismo
9.
FASEB J ; 37(3): e22820, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36801982

RESUMEN

Epidemiological studies suggest that fetal growth restriction (FGR) caused by gestational cholestasis is associated with elevated serum cholic acid (CA). Here, we explore the mechanism by which CA induces FGR. Pregnant mice except controls were orally administered with CA daily from gestational day 13 (GD13) to GD17. Results found that CA exposure decreased fetal weight and crown-rump length, and increased the incidence of FGR in a dose-dependent manner. Furthermore, CA caused placental glucocorticoid (GC) barrier dysfunction via down-regulating the protein but not the mRNA level of placental 11ß-Hydroxysteroid dehydrogenase-2 (11ß-HSD2). Additionally, CA activated placental GCN2/eIF2α pathway. GCN2iB, an inhibitor of GCN2, significantly inhibited CA-induced down-regulation of 11ß-HSD2 protein. We further found that CA caused excessive reactive oxygen species (ROS) production and oxidative stress in mouse placentas and human trophoblasts. NAC significantly rescued CA-induced placental barrier dysfunction by inhibiting activation of GCN2/eIF2α pathway and subsequent down-regulation of 11ß-HSD2 protein in placental trophoblasts. Importantly, NAC rescued CA-induced FGR in mice. Overall, our results suggest that CA exposure during late pregnancy induces placental GC barrier dysfunction and subsequent FGR may be via ROS-mediated placental GCN2/eIF2α activation. This study provides valuable insight for understanding the mechanism of cholestasis-induced placental dysfunction and subsequent FGR.


Asunto(s)
Enfermedades Placentarias , Placenta , Embarazo , Femenino , Ratones , Humanos , Animales , Placenta/metabolismo , Especies Reactivas de Oxígeno/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Retardo del Crecimiento Fetal/inducido químicamente , Factor 2 Eucariótico de Iniciación/metabolismo , Enfermedades Placentarias/metabolismo
10.
Curr Hypertens Rep ; 24(6): 157-172, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35254588

RESUMEN

PURPOSE OF REVIEW: Preeclampsia complicates 5-10% of all pregnancies and is a leading cause of maternal and perinatal mortality and morbidity. The placenta plays a pivotal role in determining pregnancy outcome by supplying the fetus with oxygen and nutrients and by synthesizing hormones. Placental function is highly dependent on energy supplied by mitochondria. It is well-known that preeclampsia is originated from placental dysfunction, although the etiology of it remains elusive. RECENT FINDINGS: During the last three decades, substantial evidence suggests that mitochondrial abnormality is a major contributor to placental dysfunction. In addition, mitochondrial damage caused by circulating bioactive factors released from the placenta may cause endothelial dysfunction and subsequent elevation in maternal blood pressure. In this review, we summarize the current knowledge of mitochondrial abnormality in the pathogenesis of preeclampsia and discuss therapeutic approaches targeting mitochondria for treatment of preeclampsia.


Asunto(s)
Hipertensión , Enfermedades Placentarias , Preeclampsia , Femenino , Humanos , Hipertensión/complicaciones , Mitocondrias , Placenta , Enfermedades Placentarias/metabolismo , Enfermedades Placentarias/patología , Preeclampsia/etiología , Embarazo
11.
J Obstet Gynaecol Res ; 48(4): 920-929, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35104920

RESUMEN

AIM: The aim of this study was to determine whether there was a difference in placental metabolite profiles between patients with fetal growth restriction (FGR) and healthy controls. METHODS: The study included 10 patients with FGR diagnosis with 14 healthy controls with both matched maternal age and body mass index. 1 H HR-MAS NMR spectroscopy data obtained from placental tissue samples of patients with FGR and healthy control group were analyzed with bioinformatics methods. The obtained results of metabolite levels were further validated with the internal standard (IS) quantification method. RESULTS: Principal component analysis (PCA) and the partial least squares discriminant analysis (PLS-DA) score plots obtained with the multivariate statistical analysis of preprocessed spectral data shows a separation between the samples from patients with FGR and healthy controls. Bioinformatics analysis results suggest that the placental levels of lactate, glutamine, glycerophosphocholine, phosphocholine, taurine, and myoinositol are increased in patients with FGR compared to the healthy controls. CONCLUSIONS: Placental metabolic dysfunctions are a common occurrence in FGR.


Asunto(s)
Retardo del Crecimiento Fetal , Enfermedades Placentarias , Femenino , Retardo del Crecimiento Fetal/diagnóstico , Humanos , Edad Materna , Metabolómica , Placenta/metabolismo , Enfermedades Placentarias/metabolismo , Embarazo
12.
Am J Obstet Gynecol ; 226(2S): S1019-S1034, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33096092

RESUMEN

Preeclampsia is a devastating medical complication of pregnancy that can lead to significant maternal and fetal morbidity and mortality. It is currently believed that there is abnormal placentation in as early as the first trimester in women destined to develop preeclampsia. Although the etiology of the abnormal placentation is being debated, numerous epidemiologic and experimental studies suggest that imbalances in circulating angiogenic factors released from the placenta are responsible for the maternal signs and symptoms of preeclampsia. In particular, circulating levels of soluble fms-like tyrosine kinase 1, an antiangiogenic factor, are markedly increased in women with preeclampsia, whereas free levels of its ligand, placental, growth factor are markedly diminished. Alterations in these angiogenic factors precede the onset of clinical signs of preeclampsia and correlate with disease severity. Recently, the availability of automated assays for the measurement of angiogenic biomarkers in the plasma, serum, and urine has helped investigators worldwide to demonstrate a key role for these factors in the clinical diagnosis and prediction of preeclampsia. Numerous studies have reported that circulating angiogenic biomarkers have a very high negative predictive value to rule out clinical disease among women with suspected preeclampsia. These blood-based biomarkers have provided a valuable tool to clinicians to accelerate the time to clinical diagnosis and minimize maternal adverse outcomes in women with preeclampsia. Angiogenic biomarkers have also been useful to elucidate the pathogenesis of related disorders of abnormal placentation such as intrauterine growth restriction, intrauterine fetal death, twin-to-twin transfusion syndrome, and fetal hydrops. In summary, the discovery and characterization of angiogenic proteins of placental origin have provided clinicians a noninvasive blood-based tool to monitor placental function and health and for early detection of disorders of placentation. Uncovering the mechanisms of altered angiogenic factors in preeclampsia and related disorders of placentation may provide insights into novel preventive and therapeutic options.


Asunto(s)
Factor de Crecimiento Placentario/sangre , Preeclampsia/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Biomarcadores/sangre , Displasia Broncopulmonar/sangre , Enfermedades Cardiovasculares/sangre , Femenino , Muerte Fetal , Transfusión Feto-Fetal , Fibrina/metabolismo , Humanos , Hidropesía Fetal/sangre , Enfermedades Placentarias/metabolismo , Factor de Crecimiento Placentario/orina , Placentación , Preeclampsia/diagnóstico , Embarazo , Pronóstico , Trastornos Puerperales/sangre , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/sangre
13.
Am J Surg Pathol ; 46(2): 258-267, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34799484

RESUMEN

Bizarre (atypical/symplastic) cells have been described in various gynecologic normal tissues and benign neoplasms. This type of bizarre cytologic change is usually an incidental finding and is regarded as a benign process. We describe 17 cases of bizarre chorionic-type trophoblast in second-trimester and third-trimester placentas that created concern for an underlying/undersampled or incipient intraplacental trophoblastic neoplasm, predominantly found in intervillous trophoblastic islands (11/17), placental septae (6/17), chorionic plate (1/17), and/or the chorion layer of fetal membranes (2/17). The bizarre trophoblastic cells exhibited sheet-like or nested architecture, had a multifocal/patchy distribution, and/or were present as individual cells within hyaline stroma; they were characterized by large nuclei with smudgy chromatin and occasional intranuclear pseudoinclusions. The degree of atypia was classified as mild (0/17), moderate (3/17), or severe (14/17). Mitotic figures and necrosis were not identified. A dual immunohistochemical stain for trophoblast (hydroxyl-delta-5-steroid dehydrogenase) and a proliferation marker (Ki-67), performed in 15 cases, demonstrated 0% to very low proliferative activity within the bizarre trophoblast (0% to 2% [10/15], 3% to 8% [5/15]). Immunohistochemical stains for fumarate hydratase showed intact/retained expression in the bizarre cells in 7 of 7 cases. Clinical follow-up ranged from 1 to 45 months, and all patients were alive and well without subsequent evidence of a gestational trophoblastic or other neoplasms. We conclude that bizarre chorionic-type trophoblast in second-trimester or third-trimester placentas have the potential to mimic an intraplacental trophoblastic neoplasm but are likely a benign degenerative change. This study expands the spectrum of bizarre cells that occur in the gynecologic tract.


Asunto(s)
Enfermedades Placentarias/patología , Neoplasias Trofoblásticas/patología , Trofoblastos/patología , Neoplasias Uterinas/patología , Adolescente , Adulto , Biopsia , Diagnóstico Diferencial , Femenino , Fumarato Hidratasa/análisis , Humanos , Inmunohistoquímica , Antígeno Ki-67/análisis , Persona de Mediana Edad , Complejos Multienzimáticos/análisis , Enfermedades Placentarias/metabolismo , Valor Predictivo de las Pruebas , Embarazo , Segundo Trimestre del Embarazo , Tercer Trimestre del Embarazo , Progesterona Reductasa/análisis , Esteroide Isomerasas/análisis , Neoplasias Trofoblásticas/química , Trofoblastos/química , Estados Unidos , Neoplasias Uterinas/química , Adulto Joven
14.
Sci Rep ; 11(1): 20670, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667209

RESUMEN

Nuclear Receptor Subfamily 4 Group A Member 2 (NR4A2) transcripts are elevated in the circulation of individuals whose pregnancies are complicated by preterm fetal growth restriction (FGR). In this paper, we show that the cases with preeclampsia (PE) have increased circulating NR4A2 transcripts compared to those with normotensive FGR. We aimed to establish whether the dysfunctional placenta mirrors the increase in NR4A2 transcripts and further, to uncover the function of placental NR4A2. NR4A2 expression was detected in preterm and term placental tissue; expressed higher at term. NR4A2 mRNA expression and protein were not altered in placentas from preterm FGR or PE pregnancies. Hypoxia (1% O2 compared to 8% O2) significantly reduced cytotrophoblast NR4A2 mRNA expression, but not placental explant NR4A2 expression. Silencing cytotrophoblast NR4A2 expression under hypoxia (via short interfering (si)RNAs) did not alter angiogenic Placental Growth Factor, nor anti-angiogenic sFlt-1 mRNA expression or protein secretion, but increased expression of cellular antioxidant, oxidative stress, inflammatory, and growth genes. NR4A2 expression was also not altered in a model of tumour necrosis factor-α-induced endothelial dysfunction, or with pravastatin treatment. Further studies are required to identify the origin of the circulating transcripts in pathological pregnancies, and investigate the function of placental NR4A2.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Factor de Crecimiento Placentario/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Trofoblastos/metabolismo , Adulto , Antioxidantes/metabolismo , Femenino , Humanos , Hipoxia/metabolismo , Inflamación/metabolismo , Masculino , Estrés Oxidativo/fisiología , Enfermedades Placentarias/metabolismo , Embarazo , ARN Mensajero/metabolismo
15.
Int J Mol Sci ; 22(15)2021 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-34360662

RESUMEN

A well-functioning placenta is crucial for normal gestation and regulates the nutrient, gas, and waste exchanges between the maternal and fetal circulations and is an important endocrine organ producing hormones that regulate both the maternal and fetal physiologies during pregnancy. Placental insufficiency is implicated in spontaneous preterm birth (SPTB). We proposed that deficits in the capacity of the placenta to maintain bioenergetic and metabolic stability during pregnancy may ultimately result in SPTB. To explore our hypothesis, we performed a RNA-seq study in male and female placentas from women with SPTB (<36 weeks gestation) compared to normal pregnancies (≥38 weeks gestation) to assess the alterations in the gene expression profiles. We focused exclusively on Black women (cases and controls), who are at the highest risk of SPTB. Six hundred and seventy differentially expressed genes were identified in male SPTB placentas. Among them, 313 and 357 transcripts were increased and decreased, respectively. In contrast, only 61 differentially expressed genes were identified in female SPTB placenta. The ingenuity pathway analysis showed alterations in the genes and canonical pathways critical for regulating inflammation, oxidative stress, detoxification, mitochondrial function, energy metabolism, and the extracellular matrix. Many upstream regulators and master regulators important for nutrient-sensing and metabolism were also altered in SPTB placentas, including the PI3K complex, TGFB1/SMADs, SMARCA4, TP63, CDKN2A, BRCA1, and NFAT. The transcriptome was integrated with published human placental metabolome to assess the interactions of altered genes and metabolites. Collectively, significant and biologically relevant alterations in the transcriptome were identified in SPTB placentas with fetal sex disparities. Altered energy metabolism, mitochondrial function, inflammation, and detoxification may underly the mechanisms of placental dysfunction in SPTB.


Asunto(s)
Metabolismo Energético , Inflamación/patología , Enfermedades Placentarias/patología , Placenta/patología , Nacimiento Prematuro/patología , Transcriptoma , Adulto , Femenino , Edad Gestacional , Humanos , Recién Nacido , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Placenta/inmunología , Placenta/metabolismo , Enfermedades Placentarias/genética , Enfermedades Placentarias/inmunología , Enfermedades Placentarias/metabolismo , Embarazo , Nacimiento Prematuro/genética , Nacimiento Prematuro/inmunología , Nacimiento Prematuro/metabolismo , Factores Sexuales
16.
Int J Mol Sci ; 22(14)2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34299087

RESUMEN

Biomarkers for placental dysfunction are currently lacking. We recently identified SPINT1 as a novel biomarker; SPINT2 is a functionally related placental protease inhibitor. This study aimed to characterise SPINT2 expression in placental insufficiency. Circulating SPINT2 was assessed in three prospective cohorts, collected at the following: (1) term delivery (n = 227), (2) 36 weeks (n = 364), and (3) 24-34 weeks' (n = 294) gestation. SPINT2 was also measured in the plasma and placentas of women with established placental disease at preterm (<34 weeks) delivery. Using first-trimester human trophoblast stem cells, SPINT2 expression was assessed in hypoxia/normoxia (1% vs. 8% O2), and following inflammatory cytokine treatment (TNFα, IL-6). Placental SPINT2 mRNA was measured in a rat model of late-gestational foetal growth restriction. At 36 weeks, circulating SPINT2 was elevated in patients who later developed preeclampsia (p = 0.028; median = 2233 pg/mL vs. controls, median = 1644 pg/mL), or delivered a small-for-gestational-age infant (p = 0.002; median = 2109 pg/mL vs. controls, median = 1614 pg/mL). SPINT2 was elevated in the placentas of patients who required delivery for preterm preeclampsia (p = 0.025). Though inflammatory cytokines had no effect, hypoxia increased SPINT2 in cytotrophoblast stem cells, and its expression was elevated in the placental labyrinth of growth-restricted rats. These findings suggest elevated SPINT2 is associated with placental insufficiency.


Asunto(s)
Biomarcadores/metabolismo , Retardo del Crecimiento Fetal/diagnóstico , Glicoproteínas de Membrana/metabolismo , Enfermedades Placentarias/diagnóstico , Placenta/patología , Preeclampsia/diagnóstico , Trofoblastos/patología , Adolescente , Femenino , Retardo del Crecimiento Fetal/metabolismo , Humanos , Recién Nacido , Recién Nacido Pequeño para la Edad Gestacional , Estudios Longitudinales , Placenta/metabolismo , Enfermedades Placentarias/metabolismo , Preeclampsia/metabolismo , Embarazo , Estudios Prospectivos , Trofoblastos/metabolismo
17.
Adv Protein Chem Struct Biol ; 126: 195-225, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34090615

RESUMEN

Autophagy and apoptosis are catabolic pathways essential for homeostasis. They play a crucial role for normal placental and fetal development. These cell death mechanisms are exaggerated in placental disorders such as preeclampsia, intrauterine growth restriction (IUGR) and gestational diabetes mellitus (GDM). Apoptosis is widely studied, highly controlled and regulated whereas; autophagy is an orderly degradation and recycling of the cellular components. Cellular senescence may be initiated by a variety of stimuli, including hypoxia, oxidative stress, reduction in survival signals and nutrition deprivation. Apoptosis is regulated by two types of pathways intrinsic and extrinsic. Extrinsic pathway is initiated by apoptosis inducing cells such as macrophages, natural killer cells whereas; intrinsic pathway is initiated in response to DNA damage, cell injury and lack of oxygen. In autophagy, the cell or organelles undergo lysosomal degradation. Placental apoptosis increases as the gestation progresses while autophagy plays a role in trophoblast differentiation and invasion. In pregnancy disorders like preeclampsia and IUGR, proapoptotic markers such as caspase 3, 8, BAX are higher and antiapoptotic markers like Bcl-2 are lower. In GDM, apoptotic markers are reduced resulting in increased placental mass and fetal macrosomia. Apoptosis in the pathological pregnancies is also influenced by the reduced levels of micronutrients and long chain polyunsaturated fatty acids resulting in disturbed placental biology. This chapter describes the role of various key molecular events involved in cellular senescence and the various factors influencing them. This will help identify future therapeutic strategies for better management of these processes.


Asunto(s)
Apoptosis , Senescencia Celular , Daño del ADN , Estrés Oxidativo , Enfermedades Placentarias/metabolismo , Trofoblastos/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Hipoxia de la Célula , Femenino , Humanos , Enfermedades Placentarias/patología , Enfermedades Placentarias/terapia , Embarazo
18.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669156

RESUMEN

Pregnancy complications are a major cause of fetal and maternal morbidity and mortality in humans. The majority of pregnancy complications initiate due to abnormal placental development and function. During the last decade, the role of microRNAs (miRNAs) in regulating placental and fetal development has become evident. Dysregulation of miRNAs in the placenta not only affects placental development and function, but these miRNAs can also be exported to both maternal and fetal compartments and affect maternal physiology and fetal growth and development. Due to their differential expression in the placenta and maternal circulation during pregnancy complications, miRNAs can be used as diagnostic biomarkers. However, the differential expression of a miRNA in the placenta may not always be reflected in maternal circulation, which makes it difficult to find a reliable biomarker for placental dysfunction. In this review, we provide an overview of differentially expressed miRNAs in the placenta and/or maternal circulation during preeclampsia (PE) and intrauterine growth restriction (IUGR), which can potentially serve as biomarkers for prediction or diagnosis of pregnancy complications. Using different bioinformatics tools, we also identified potential target genes of miRNAs associated with PE and IUGR, and the role of miRNA-mRNA networks in the regulation of important signaling pathways and biological processes.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , MicroARNs/metabolismo , Enfermedades Placentarias/metabolismo , Preeclampsia/metabolismo , Transcriptoma/genética , Biomarcadores/sangre , Femenino , Retardo del Crecimiento Fetal/genética , Ontología de Genes , Humanos , MicroARNs/genética , Enfermedades Placentarias/genética , Placentación/genética , Preeclampsia/genética , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/genética
19.
Reproduction ; 161(6): 603-621, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33780349

RESUMEN

Cervical remodeling is a critical component in both term and preterm labor in eutherian mammals. However, the molecular mechanisms underlying cervical remodeling remain poorly understood in the mare. The current study compared the transcriptome of the equine cervix (cervical mucosa (CM) and stroma (CS)) during placentitis (placentitis group, n = 5) and normal prepartum mares (prepartum group, n = 3) to normal pregnant mares (control group, n = 4). Transcriptome analysis identified differentially expressed genes (DEGs) during placentitis (5310 in CM and 907 in CS) and during the normal prepartum period (189 in CM and 78 in CS). Our study revealed that cervical remodeling during placentitis was dominated by inflammatory signaling as reflected by the overrepresented toll-like receptor signaling, interleukin signaling, T cell activation, and B cell activation pathways. These pathways were accompanied by upregulation of several proteases, including matrix metalloproteinases (MMP1, MMP2, and MMP9), cathepsins (CTSB, CTSC, and CTSD) and a disintegrin and metalloproteinase with thrombospondin type 1 motifs (ADAMTS1, ADAMTS4, and ADAMTS5), which are crucial for degradation of cervical collagens during remodeling. Cervical remodeling during placentitis was also associated with upregulation of water channel-related transcripts (AQP9 and RLN), angiogenesis-related transcripts (NOS3, ENG1, THBS1, and RAC2), and aggrecan (ACAN), a hydrophilic glucosaminoglycan, with subsequent cervical hydration. The normal prepartum cervix was associated with upregulation of ADAMTS1, ADAMTS4, NOS3 and THBS1, which might reflect an early stage of cervical remodeling taking place in preparation for labor. In conclusion, our findings revealed the possible key regulators and mechanisms underlying equine cervical remodeling during placentitis and the normal prepartum period.


Asunto(s)
Cuello del Útero/fisiopatología , Regulación de la Expresión Génica , Enfermedades de los Caballos/metabolismo , Enfermedades Placentarias/veterinaria , Placenta/metabolismo , Transcriptoma , Animales , Femenino , Enfermedades de los Caballos/genética , Enfermedades de los Caballos/patología , Caballos , Enfermedades Placentarias/genética , Enfermedades Placentarias/metabolismo , Enfermedades Placentarias/patología , Embarazo
20.
Placenta ; 116: 2-11, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33663810

RESUMEN

Despite the indispensable role of the placenta in the successful course of pregnancy, regulation of its dynamic transcriptome is still underexplored. The purpose of this literature review was to give an overview and draw attention to the contribution of genetic variation in shaping the human placental gene expression. Studies of placental transcriptome shaped by chromosomal variants are limited and may be confounded by cellular mosaicism and somatic genomic rearrangements. Even in relatively simple cases, such as aneuploidies, the placental transcriptome appears to differ from the assumed systematically increased transcript levels of the involved chromosomes. Single nucleotide variants modulating placental gene expression referred to as expression quantitative trait loci (eQTLs) have been analyzed only in ten candidate gene and three genome-wide association studies (GWAS). The latter identified 417 confident placental eGenes, supported by at least two independent studies. Functional profiling of eGenes highlighted biological pathways important in pregnancy, such as immune response or transmembrane transport activity. A fraction of placental eQTLs (1-3%) co-localize with GWAS loci for adult disorders (metabolic, immunological, neurological), suggesting a co-contributory role of the placenta in the developmental programming of health. Some placental eQTLs have been identified as risk factors for adverse pregnancy outcomes, such as rs4769613 (C > T), located near the FLT1 gene and confidently associated with preeclampsia. More studies are needed to map genetic variants shaping gene expression in different placental cell types across three trimesters in normal and complicated gestations and to clarify to what extent these heritable factors contribute to maternal and offspring disease risks.


Asunto(s)
Enfermedades Placentarias/metabolismo , Placenta/metabolismo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Transcriptoma , Femenino , Perfilación de la Expresión Génica , Humanos , Enfermedades Placentarias/genética , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...