bims-placeb Biomed News
on Placental cell biology
Issue of 2025–11–30
sixteen papers selected by
Carlos M Guardia, National Institute of Environmental Health Sciences



  1. bioRxiv. 2025 Oct 08. pii: 2025.10.08.681177. [Epub ahead of print]
      The expansion and differentiation of trophoblast stem (TS) cells are critical for defining fundamental properties of the placenta. Specialized trophoblast cells exit the placenta and enter and transform the uterus, including restructuring uterine spiral arteries. In the human these cells are referred to as extravillous trophoblast (EVT) cells, whereas in the rat they are termed invasive trophoblast cells. Mechanisms governing invasive trophoblast cell differentiation remain poorly understood. We investigated peroxisome proliferator-activated receptor gamma (PPARG) as a potential regulator of EVT/invasive trophoblast cell development. In first trimester human placentas, PPARG transcripts and protein were detected in the EVT cell column and increased in amount as human TS cells differentiated into EVT cells. Disruption of PPARG impaired differentiation of human TS cells into EVT cells. Invasive trophoblast cells of the rat placentation site similarly expressed PPARG. Conditional inactivation of PPARG within invasive trophoblast cells of the rat placenta was used to assess the in vivo role of PPARG on the uterine-placental interface. PPARG was established as an essential cell-autonomous regulator of the invasive trophoblast cell lineage with profound effects on placentation. In conclusion, PPARG is a conserved regulator of placentation and is essential for directing trophoblast cell-guided uterine transformation.
    DOI:  https://doi.org/10.1101/2025.10.08.681177
  2. Sci Rep. 2025 Nov 26.
      The majority of pregnancy loss in ruminants occurs during the first two months of gestation, and a failure in placenta development is a major cause of pregnancy loss in cattle after day 20. Gaining a cell-type level understanding of normal placental development is essential for uncovering how this critical organ, responsible for nutrient exchange, gas transfer, and waste removal, fails during pregnancy loss. This study integrated single-cell RNA sequencing (scRNA-seq) from sheep and cattle during early placental development. Nineteen distinct cell populations were identified across species, with mesenchymal, epithelial, and trophoblast cells showing largely conserved expression profiles. Interestingly, two trophoblast clusters were unique to cattle, with one expressing IFNT2 (uninucleate) and another expressing CSH2 and PAG17 (binucleate). Genes associated with epithelial-to-mesenchymal transition (EMT), such as SNAI1, SNAI2, ZEB1, VIM, CDH1, and CLDN4, showed dynamic and prominent expression patterns in trophoblasts. Pseudotime and cell-cell signaling analyses supported the occurrence of EMT in uninucleate trophoblasts. Gene ontology comparisons revealed similarities between ruminant and human extravillous trophoblasts, suggesting conserved EMT across placental types. Collectively, these findings highlight EMT as a potentially critical process in early ruminant placentation.
    Keywords:  Comparative genomics; Placentation; Ruminants; Single-cell transcriptomics
    DOI:  https://doi.org/10.1038/s41598-025-29895-2
  3. J Virol. 2025 Nov 25. e0161025
      Human cytomegalovirus (HCMV) is the leading infectious cause of congenital disease, but the mechanisms governing vertical transmission remain poorly defined. Murine cytomegalovirus (MCMV) infection in mice is a widely used model to study pathogenesis in vivo. However, the mouse model cannot be used to study congenital infection as viral transmission from mother to fetus in utero does not occur. In this study, we investigated tissue-specific features of the murine materno-fetal barrier that may restrict viral infection. Following high-dose intravenous challenge of wild-type and severely immunocompromised pregnant mice, MCMV replicated efficiently in the maternal liver but infected only a few cells in the placenta, suggesting an intrinsic resistance. Ex vivo analyses of primary placental cells, trophoblast stem cells, and a trophoblast cell line revealed a low susceptibility to MCMV infection compared to other permissive cell types. The resistance of trophoblast cells to MCMV infection correlated with the absence of neuropilin-1 (NRP1), a cellular receptor required for efficient infection of several cell types. Enforced expression of NRP1 in trophoblast cells increased their susceptibility to MCMV infection and replication, suggesting that the resistance of trophoblast cells to MCMV infection is caused by the lack of a critical receptor. These findings further suggest that, in addition to immune-mediated restrictions, cell-intrinsic resistance limits MCMV infection and transmission at the murine materno-fetal interface.IMPORTANCECongenital cytomegalovirus (CMV) infection is a major cause of developmental disabilities in newborns, yet the biological factors that influence transmission from mother to fetus remain unclear. In this study, we demonstrate that trophoblast cells of the murine placenta are naturally resistant to CMV infection as they lack expression of a host protein, neuropilin-1 (NRP1), that the virus requires for entry. By introducing this protein into resistant cells, we demonstrated that susceptibility to infection can be reinstated, indicating that the absence of NRP1 plays a key protective role at the materno-fetal barrier. These results provide insight into why mice rarely transmit CMV to their offspring and how species-specific differences in placental biology shape susceptibility. Understanding these mechanisms will aid in refining animal models and may help identify new targets to prevent congenital infection in humans.
    Keywords:  CMV; MCMV; NRP1; congenital; cytomegalovirus; interferon; neuropilin-1; placenta; trophoblast
    DOI:  https://doi.org/10.1128/jvi.01610-25
  4. Nat Rev Endocrinol. 2025 Nov 25.
      The intricate processes of feto-placental development are regulated by both genetics and the nutritional environment. The placenta mediates nutrient transfer to the fetus and waste transfer to the maternal circulation to facilitate proper fetal cell fate specification. Disruptions in these fate-regulatory programmes can lead to maternal and fetal complications. We explore how metabolic-epigenetic mechanisms programme feto-placental development, discussing the roles of metabolic sensing mechanisms. This Review covers the roles of nutrient metabolite-dependent protein modifications, lipid signalling, steroid signalling and oxygen signalling in regulating particular phases and lineages of feto-placental development. Understanding these environmental-genetic interactions is crucial for elucidating the limits of developmental plasticity, the pathogenesis of pregnancy-related disorders and the potential therapeutic windows we can leverage to improve maternal and fetal outcomes.
    DOI:  https://doi.org/10.1038/s41574-025-01206-9
  5. FASEB J. 2025 Nov 30. 39(22): e71254
      High-altitude pregnancy presents the complex physiological challenge of fulfilling maternal, placental, and fetal metabolic demands under chronic ambient hypoxia. Highland Andeans exhibit signs of adaptation to high-altitude hypoxia, showing relative protection against altitude-associated fetal growth restriction (FGR) and the positive selection of metabolic genes linked to placental mitochondrial capacity. Not all infants are protected, with both FGR and preeclampsia occurring among highland-resident Andeans. In Andeans, placental metabolic dysfunction is evident. By integrating metabolomic studies of maternal-placental-fetal triads with adaptive genetic signals in the fetal genome, we sought to identify adaptive and maladaptive placental metabolic phenotypes in highland Andeans (La Paz, Bolivia; 3850 m), including normotensive and preeclamptic pregnancies. Widespread differences in metabolite abundance were evident between normotensive and preeclamptic pregnancy across maternal, placental, and fetal compartments. Preeclampsia was characterized by a pronounced accumulation of fatty acid derivatives, specifically medium and long-chain acylcarnitines; these were also associated with low birth weight. Genotype-phenotype association analyses revealed novel links between putatively adaptive fetal haplotypes and placental metabolite abundance. Carriers of specific adaptive fetal haplotypes comprising genes linked to lipid metabolism had a greater abundance of placental short-chain acetyl-carnitine alongside decreased levels of linolenic acid (CPT2/LRP8), lower levels of the medium-chain octanoylcarnitine (EXOC4), and greater abundance of free carnitine (LIPG). Collectively, our study reveals a distinct metabolic phenotype in Andean preeclampsia characterized by incomplete fatty acid oxidation and highlights novel links between putatively adaptive fetal haplotypes and healthy placental metabolic phenotypes.
    Keywords:  adaptation; hypertensive disorders of pregnancy; hypoxia; metabolome
    DOI:  https://doi.org/10.1096/fj.202502590R
  6. Cell Death Dis. 2025 Nov 24.
      Preeclampsia is a severe pregnancy complication marked by impaired trophoblast function and abnormal placental development, leading to significant maternal and fetal morbidity. FK506-binding protein-like (FKBPL) has been identified as a potential biomarker as it is significantly downregulated in early pregnancy stages of women who progress to develop preeclampsia. However, editing the Fkbpl gene in trophoblast cells to create a model of preeclampsia using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology is challenging due to inefficient delivery, leading to low editing efficiency and reduced cell viability. To address these challenges, we developed a cost-effective and minimally invasive mechanoporation system using micro-engineered filters to deliver CRISPR/Cas9 plasmid DNA (pDNA) targeting the Fkbpl gene into trophoblast cells. This approach successfully generated cell lines with a 38% knockout (K/O) of Fkbpl expression, significantly reducing cell migration (wildtype (WT): 28.77% ± 4.7 vs. 38% K/O: 4.95% ± 0.8, wound closure, **p < 0.01) and proliferation (WT: 1.26 ± 0.06 vs. 38% K/O: 0.81 ± 0.01, ****p < 0.0001). Lower Fkbpl-K/O efficiency of 17% showed a similar reduction in cell proliferation as the 38% K/O clone. Although a full Fkbpl-K/O in the ACH-3P first-trimester trophoblast cell line was not achieved, the partial K/O provided valuable insights into Fkbpl's role in trophoblast function relevant to preeclampsia pathogenesis. Moreover, treatment with mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEVs) or MSC-sEVs did not restore migratory capacity in Fkbpl-deficient cells (p = 0.14). MSC-sEVs increased proliferation in WT ACH-3P cells at 1 µg (p < 0.05) and 2 µg (p < 0.01) doses, however, were not effective in either 17% or 38% Fkbpl-K/O clones, suggesting that FKBPL is an important mechanism of MSC-sEV-mediated therapeutic effect in trophoblasts in the context of preeclampsia. This study advances gene-editing techniques in placental biology and proposes new therapeutic strategies and mechanisms for pregnancy-related complications.A Schematic overview of CRISPR/Cas9 plasmid delivery using microfiltroporation compared to gold standard electroporation and lipofection technologies in trophoblast cells. A CRISPR/Cas9 plasmid targeting Fkbpl was delivered to the first trimester trophoblast cell line, ACH-3P. Cells were sorted according to green fluorescence protein (GFP) expression, expanded and assessed for changes in cell function using proliferation and migration assays. B Actual images of the isopore silicon nitride (SiN) microfilters used in this study and diagram of cell membrane dynamics in response to mechanoporation. This figure was created with Biorender.com. CRISPR clustered regularly interspaced short palindromic repeats, EP electroporation, MFP microfiltroporation.
    DOI:  https://doi.org/10.1038/s41419-025-08200-z
  7. J Neuroendocrinol. 2025 Nov 27. e70120
      Miscarriage, defined as spontaneous pregnancy loss before 20 weeks of gestation, affects 10-15% of pregnancies in women under 30, rising to over 50% in women over 45. Implantation failure and early placental dysfunction are major contributors, yet the precise mechanisms remain incompletely understood. Kisspeptin, encoded by KISS1, is a critical regulator of the reproductive axis and is highly expressed in the placenta, where it modulates trophoblast proliferation, migration, invasion, and vascular remodelling. Human studies indicate that reduced placental kisspeptin is associated with recurrent pregnancy loss, while circulating kisspeptin levels increase dramatically throughout gestation and may serve as a biomarker for pregnancy viability. Mouse models, including CBA/J × DBA/2 matings and Kiss1/Kiss1r knockout lines, have provided mechanistic insights, showing that loss of kisspeptin signalling impairs decidualization, trophoblast invasion, and embryo implantation. These studies also highlight kisspeptin's role in modulating maternal immune responses and in coordinating hormonal cues, including progesterone, oestrogen, and prolactin, necessary for uterine receptivity. Despite shared features of placentation signal and endocrine regulation, significant species-specific differences exist (e.g. mice lack hCG, exhibit embryo-dependent decidualization, have less invasive trophoblasts) limiting direct extrapolation to human pregnancy. This review synthesizes current evidence on kisspeptin's paracrine and endocrine roles during early gestation, emphasizing the insights gained from murine models while highlighting the translational challenges in applying these findings to human miscarriage research.
    Keywords:  kisspeptin; miscarriage; pregnancy
    DOI:  https://doi.org/10.1111/jne.70120
  8. bioRxiv. 2025 Nov 13. pii: 2025.11.12.687933. [Epub ahead of print]
      Late-onset preeclampsia (LOPE) is a major pregnancy complication characterised by hypertension and placental dysfunction, resolving only upon delivery. Here, we show that LOPE placentae undergo accelerated molecular aging, marked by telomere attrition, DNA damage and trophoblast senescence. Using primary placental tissue and trophoblast organoids, we demonstrate oxidative stress as a driver of telomere shortening and angiogenic imbalance. Inflammation did not alter placental aging trajectories. Antioxidant treatment (superoxide dismutase) preserved telomere length, reduced DNA damage and restored angiogenic balance, highlighting oxidative stress as a modifiable determinant of placental aging. We identify reduced expression of telomeric repeat-containing RNAs (TERRAs) as a molecular hallmark of LOPE, and show that antisense oligonucleotide-mediated TERRA depletion exacerbates telomere erosion and senescence. Together, these findings delineate oxidative stress and TERRA loss as mechanisms driving placental decline, establish trophoblast organoids as a tractable model of placental aging, and reveal potential therapeutic avenues for mitigating preeclampsia-associated placental dysfunction.
    DOI:  https://doi.org/10.1101/2025.11.12.687933
  9. Placenta. 2025 Nov 21. pii: S0143-4004(25)00744-1. [Epub ahead of print]174 1-8
       BACKGROUND: Placental insufficiency underlies major obstetric complications such as preeclampsia and fetal growth restriction (FGR). The reduced uterine perfusion pressure (RUPP) model is widely used to mimic PE and FGR; however, its impact on placental structure, immune cell regulation, and vascular remodeling in the mesometrial triangle remains poorly examined. This study investigates these processes to clarify the model's relevance for human placenta dysfunction pathology.
    METHODS: Maternal, fetal, and placental parameters were assessed in pregnant rats at gestational day (GD) 14, 16, 18, and 19, comparing normal pregnant and RUPP groups. Placental morphology was assessed using hematoxylin and eosin staining. Within the mesometrial triangle, uterine natural killer (uNK) cell distribution was evaluated using ANK61 staining, while trophoblast invasion and spiral artery remodeling were assessed by pan-cytokeratin and α-SMA staining, respectively.
    RESULTS: Placental macrostructural architecture was preserved in RUPP. However, uNK cell migration from the mesometrial triangle was impaired, with higher ANK61-positive cell presence persisting at GD19. This was accompanied by a reduction in trophoblast invasion depth. Spiral artery remodeling was impaired in RUPP placentas, with a greater proportion of unremodeled vessels and fewer highly remodeled arteries by GD19.
    CONCLUSION: While gross placental structure remains intact in the RUPP model, the key functional adaptations uNK cell migration, trophoblast invasion, and vascular remodeling, are impaired. These findings support the RUPP as a relevant model to study the pathophysiological mechanisms of placental insufficiency. Future research should focus on temporal molecular profiling to elucidate the mechanisms behind impaired uNK cell retention and shallow trophoblast invasion.
    Keywords:  Placental insufficiency; Placental vascular development; RUPP model; Spiral artery remodeling; Trophoblast invasion; uNK cells
    DOI:  https://doi.org/10.1016/j.placenta.2025.11.008
  10. bioRxiv. 2025 Oct 23. pii: 2025.10.22.683996. [Epub ahead of print]
    Birth Defects Research Laboratory (BDRL)
       Objectives: Metabolic demands of the developing conceptus are highly dynamic during pregnancy. While placental metabolism has been well described at term and in cell lines, changes in the placental metabolome during development remains understudied. We investigated the placental metabolome, metabolite trajectories, and altered pathways across trimesters in normal human pregnancy by integrating metabolomic and transcriptomic data.
    Methods: Targeted aqueous metabolomic profiling of 372 metabolites was conducted on placental biopsies from samples collected in the first (n=12), second (n=13), and third (n=11) trimesters of normal pregnancy using liquid chromatography-tandem mass spectrometry. Robust linear models identified differentially abundant metabolites across trimesters in models adjusted for fetal sex and total protein. We conducted pathway analysis using a human metabolic reconstruction. To further aid in biological interpretation, we leveraged publicly available transcriptomics data to conduct pathway-level multi-omic integration throughout gestation.
    Results: Samples clustered by trimester in principal component analysis and we identified 5 metabolite trajectories. Out of 193 detectable metabolites, 149 (77%) differed by trimester (FDR<0.05). Using pathway-level multi-omic integration, pathways involved in extracellular transport, and pyruvate, amino acid, NAD, and membrane lipid metabolism are up-regulated in the second trimester compared to the first. In the late third trimester, pathways involved in amino acid metabolism, redox balance, mitochondrial transport, and biomolecule synthesis were down-regulated compared to second trimester.
    Conclusions: Placental metabolite abundances change substantially across gestation and integration with metabolic gene expression provides insight into dynamic metabolic function during pregnancy. Observed pathway-level changes potentially reflect the metabolic response to invading maternal circulation in the first-to-second trimester transition, as well as changing maternal and fetal metabolic requirements. Differences observed at term may reflect placental senescence and preparation for parturition. These data can inform other molecular analyses of the placenta by providing enhanced resolution of metabolic changes across pregnancy.
    DOI:  https://doi.org/10.1101/2025.10.22.683996
  11. Front Cell Dev Biol. 2025 ;13 1678878
       Objective: We investigated the role of neural Wiskott-Aldrich syndrome protein (N-WASP) in preeclampsia (PE), focusing on its regulatory impact on trophoblast syncytialization.
    Methods: We analyzed placental samples from patients with PE (n = 30) and controls (n = 35) using RNA extraction, quantitative real-time polymerase chain reaction, Western blot, and immunohistochemistry. BeWo cell lines were used to model trophoblast fusion under forskolin stimulation. We explored N-WASP's role in trophoblast cell behavior using gene knockdown and overexpression experiments. Using bioinformatics analyses and molecular docking studies, we elucidated the interaction between N-WASP and associated pathways. These findings were validated in vivo using an L-NAME PE rat model.
    Results: N-WASP expression was significantly reduced in PE placentas, correlating positively with syncytin-2 and GCM1 levels. In BeWo cells, N-WASP promoted syncytialization by activating the FAK/β-catenin pathway, causing increased nuclear β-catenin translocation, glial cells missing 1 expression, and syncytin-2 transcription. Mechanistically, N-WASP interacted with myosin 1B causing FAK pathway activation. Restoring N-WASP expression ameliorated placental abnormalities and PE symptoms in vivo. We identified hydroxychloroquine as a potential N-WASP agonist, capable of enhancing trophoblast syncytialization in vitro using molecular docking. Treatment with hydroxychloroquine significantly improved clinical symptoms, including reducing elevated blood pressure, decreasing urinary protein levels, and normalizing serum creatinine concentrations in PE rat models.
    Conclusion: We identified N-WASP as a key regulator of trophoblast syncytialization through the FAK/β-catenin signaling pathway, influencing syncytin-2 expression. The findings reveal a novel molecular mechanism underlying PE and suggest that N-WASP is a potential therapeutic target for PE.
    Keywords:  N-WASP; preeclampsia; syncytialization; syncytin-2; trophoblast
    DOI:  https://doi.org/10.3389/fcell.2025.1678878
  12. J Physiol. 2025 Nov 23.
      Research in both humans and animals has consistently demonstrated that diverse complications during pregnancy impact on the risk of chronic diseases in the offspring. In many settings, over half of women are either overweight or living with obesity during pregnancy. This has short- and long-term impacts on offspring health. The mechanisms mediating changes in the fetal environment that may trigger developmental origins of future cardiometabolic risk in the offspring are not fully elucidated. In this study, using an established mouse model, we aimed to determine whether obesity during pregnancy causes fetal hypoxia and to explore potential underlying mechanisms. We showed that fetal hypoxia is a key component of the in utero obesogenic environment at E13.5/0.7 of gestation. Concomitantly, obese dams exhibit low iron levels, as well as higher circulating levels of hepcidin and C-reactive protein. We also showed that placental structure and efficiency are not affected by maternal obesity at E13.5, suggesting that the reduction in oxygen delivery to the fetus was not a consequence of placental dysfunction at this stage of gestation. We conclude that maternal obesity-induced iron deficiency and fetal hypoxia are important mechanisms by which obesity during pregnancy impacts offspring health. Furthermore, iron deficiency in mothers with obesity is a tractable therapeutic target for intervention that could prevent transmission of poor cardiometabolic health from mother to child. KEY POINTS: Diet-induced maternal obesity resulted in fetal, but not placental, hypoxia. Pregnant mice with obesity had lower circulating iron levels, along with dysregulation of key molecules involved in iron homeostasis, such as transferrin and the hormone hepcidin. Body weight, fat mass, circulating insulin and hepcidin levels in mothers with obesity were significantly correlated with the degree of fetal hypoxia, suggesting they were interrelated.
    Keywords:  developmental programming; fetal hypoxia; hepcidin; iron homeostasis; maternal obesity
    DOI:  https://doi.org/10.1113/JP288635
  13. Nat Commun. 2025 Nov 24. 16(1): 10358
      The placenta is a temporary organ present during pregnancy that is responsible for coordinating all aspects of pregnancy between the mother and fetus. It has a distinct epigenetic, transcriptomic, and mutational landscape with low levels of methylation, high numbers of transcribed loci, and a high mutational burden relative to somatic tissues. We present this landscape through the application of nanopore sequencing technology to provide a more comprehensive picture of female placental genomics and methylomics along with integrated haplotype-resolved transcriptomic analyses across eight trios. Whole genome sequencing of trios allows robust phasing, permitting comprehensive genome-wide investigation of parent-of-origin methylation and transcription. This enhanced view facilitates identifications of many differentially methylated regions (DMRs), both conserved and differing between individuals, as well as previously unreported imprinted genes including ILDR2 and RASA1 which are potentially important for healthy placental and fetal development.
    DOI:  https://doi.org/10.1038/s41467-025-65337-3
  14. Proc Natl Acad Sci U S A. 2025 Dec 02. 122(48): e2508432122
      The trophectoderm (TE), the first lineage specified during mammalian development, initiates implantation and gives rise to placental trophoblasts. While animal models have elucidated key conserved signaling pathways involved in early TE specification, including bone morphogenetic protein (BMP), WNT, and HIPPO, species-specific differences during early development emphasize the need for human-specific models. We previously identified VGLL1, a coactivator of TEAD transcription factors, as a human-specific placental marker. In this study, we employed a pluripotent stem cell (PSC)-based model of TE induction by BMP4 to investigate chromatin remodeling and transcriptional dynamics during TE formation. BMP4-induced chromatin accessibility changes promoted a trophoblast gene expression program, while mesoderm lineage markers were only transiently expressed upon canonical WNT activation. We found that VGLL1 was expressed downstream of key TE transcription factors (GATA2/3, TFAP2A/C) but was essential for establishment of full trophoblast identity by up-regulating the epidermal growth factor receptor (EGFR) and reinforcing GATA3 expression through positive feedback. Notably, VGLL1 enhanced canonical WNT signaling via direct regulation of WNT receptors and effectors. We also identified KDM6B, a histone demethylase that removes H3K27me3 repressive marks, as a direct VGLL1 target. KDM6B facilitated activation of bivalent promoters associated with TE markers, linking epigenetic regulation to lineage identity. Our findings establish a mechanistic framework positioning VGLL1 as a central regulator that integrates HIPPO, BMP, and WNT signaling pathways to drive establishment of human TE.
    Keywords:  VGLL1; pluripotent stem cells; trophectoderm; trophoblast stem cells
    DOI:  https://doi.org/10.1073/pnas.2508432122
  15. Sci Adv. 2025 Nov 28. 11(48): eady1693
      During mouse embryogenesis, totipotency is gradually lost, and, at the 16-cell stage, blastomeres begin to bifurcate into trophectoderm (future placenta) and inner cell mass (future fetus). Although this process is well studied, when and how blastomeres acquire the competence for lineage specification remains unclear. Here, we describe that CEBPa becomes up-regulated at the transition from the two- to the four-cell stage by NR5A2 and is also selectively expressed in the trophectoderm at the blastocyst stage. Its knockout decreases the proportion of trophectoderm cells and delays the morula to blastocyst transition. Conversely, CEBPa overexpression in mouse embryonic stem cells, used as a proxy, drives their differentiation into trophectoderm-like cells, enabling the identification of CEBPa-regulated trophectoderm-specific enhancers. A subset of these enhancers, associated with key trophectoderm-related transcription factor genes, is primed or activated in four- and eight-cell embryos. Together, our data suggest that CEBPa plays a role in the installment of trophectoderm competence before the first lineage bifurcation and in trophectoderm specification.
    DOI:  https://doi.org/10.1126/sciadv.ady1693
  16. Environ Int. 2025 Nov 20. pii: S0160-4120(25)00700-7. [Epub ahead of print]206 109949
      Prenatal exposure to phthalates, pervasive endocrine-disrupting chemicals, has been linked to child health outcomes, including prematurity and low birthweight. Placental transcriptomics data can reveal mechanisms by which environmental toxicants alter placental and fetal growth. This study aims to investigate the placental transcriptome as a mediator between prenatal maternal urinary phthalate metabolites and placental efficiency. We identified significant associations between maternal urinary concentrations of phthalate metabolites and the placental transcriptome (132 genes and 27 gene modules). Placental efficiency was modeled as the ratio of birthweight to placental weight (BW:PW) and as birthweight adjusted for placental weight (BWadj) and was also significantly associated with the placental transcriptome (460 genes and 11 gene modules). 4 genes and 9 gene modules exhibited significant mediation of the relationship between maternal urinary concentrations of phthalate metabolites and placental efficiency measures. These genes were involved in syncytialization, metabolism, DNA damage and cellular senescence, and steroid biosynthesis-processes essential to fetal growth and development because of the placenta's role in nutrient supply, hormone production, and detoxification. These findings suggest a key mediating role of the placental transcriptome in toxicological mechanisms by which phthalates may disrupt fetal growth.
    Keywords:  Birthweight; Mediation; Phthalates; Placental efficiency; Transcriptome; WGCNA
    DOI:  https://doi.org/10.1016/j.envint.2025.109949