Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.711
Filter
Add more filters

Publication year range
1.
Cell ; 187(3): 764-781.e14, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306985

ABSTRACT

Pregnancy induces dramatic metabolic changes in females; yet, the intricacies of this metabolic reprogramming remain poorly understood, especially in primates. Using cynomolgus monkeys, we constructed a comprehensive multi-tissue metabolome atlas, analyzing 273 samples from 23 maternal tissues during pregnancy. We discovered a decline in metabolic coupling between tissues as pregnancy progressed. Core metabolic pathways that were rewired during primate pregnancy included steroidogenesis, fatty acid metabolism, and arachidonic acid metabolism. Our atlas revealed 91 pregnancy-adaptive metabolites changing consistently across 23 tissues, whose roles we verified in human cell models and patient samples. Corticosterone and palmitoyl-carnitine regulated placental maturation and maternal tissue progenitors, respectively, with implications for maternal preeclampsia, diabetes, cardiac hypertrophy, and muscle and liver regeneration. Moreover, we found that corticosterone deficiency induced preeclampsia-like inflammation, indicating the atlas's potential clinical value. Overall, our multi-tissue metabolome atlas serves as a framework for elucidating the role of metabolic regulation in female health during pregnancy.


Subject(s)
Metabolomics , Pregnancy , Animals , Female , Humans , Pregnancy/metabolism , Corticosterone/metabolism , Metabolome/physiology , Placenta/metabolism , Pre-Eclampsia , Primates/metabolism
2.
Cell ; 187(1): 204-215.e14, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38070508

ABSTRACT

Mounting evidence suggests metabolism instructs stem cell fate decisions. However, how fetal metabolism changes during development and how altered maternal metabolism shapes fetal metabolism remain unexplored. We present a descriptive atlas of in vivo fetal murine metabolism during mid-to-late gestation in normal and diabetic pregnancy. Using 13C-glucose and liquid chromatography-mass spectrometry (LC-MS), we profiled the metabolism of fetal brains, hearts, livers, and placentas harvested from pregnant dams between embryonic days (E)10.5 and 18.5. Our analysis revealed metabolic features specific to a hyperglycemic environment and signatures that may denote developmental transitions during euglycemic development. We observed sorbitol accumulation in fetal tissues and altered neurotransmitter levels in fetal brains isolated from hyperglycemic dams. Tracing 13C-glucose revealed disparate fetal nutrient sourcing depending on maternal glycemic states. Regardless of glycemic state, histidine-derived metabolites accumulated in late-stage fetal tissues. Our rich dataset presents a comprehensive overview of in vivo fetal tissue metabolism and alterations due to maternal hyperglycemia.


Subject(s)
Diabetes Mellitus , Diabetes, Gestational , Fetus , Animals , Female , Mice , Pregnancy , Diabetes Mellitus/metabolism , Fetus/metabolism , Glucose/metabolism , Placenta/metabolism , Diabetes, Gestational/metabolism
3.
Cell ; 185(20): 3689-3704.e21, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179666

ABSTRACT

Regulatory landscapes drive complex developmental gene expression, but it remains unclear how their integrity is maintained when incorporating novel genes and functions during evolution. Here, we investigated how a placental mammal-specific gene, Zfp42, emerged in an ancient vertebrate topologically associated domain (TAD) without adopting or disrupting the conserved expression of its gene, Fat1. In ESCs, physical TAD partitioning separates Zfp42 and Fat1 with distinct local enhancers that drive their independent expression. This separation is driven by chromatin activity and not CTCF/cohesin. In contrast, in embryonic limbs, inactive Zfp42 shares Fat1's intact TAD without responding to active Fat1 enhancers. However, neither Fat1 enhancer-incompatibility nor nuclear envelope-attachment account for Zfp42's unresponsiveness. Rather, Zfp42's promoter is rendered inert to enhancers by context-dependent DNA methylation. Thus, diverse mechanisms enabled the integration of independent Zfp42 regulation in the Fat1 locus. Critically, such regulatory complexity appears common in evolution as, genome wide, most TADs contain multiple independently expressed genes.


Subject(s)
Chromatin , Placenta , Animals , CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Evolution, Molecular , Female , Genome , Mammals/metabolism , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Cell ; 176(1-2): 318-333.e19, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30503206

ABSTRACT

Preeclampsia is the most frequent pregnancy-related complication worldwide with no cure. While a number of molecular features have emerged, the underlying causal mechanisms behind the disorder remain obscure. Here, we find that increased complex formation between angiotensin II AT1 and bradykinin B2, two G protein-coupled receptors with opposing effects on blood vessel constriction, triggers symptoms of preeclampsia in pregnant mice. Aberrant heteromerization of AT1-B2 led to exaggerated calcium signaling and high vascular smooth muscle mechanosensitivity, which could explain the onset of preeclampsia symptoms at late-stage pregnancy as mechanical forces increase with fetal mass. AT1-B2 receptor aggregation was inhibited by beta-arrestin-mediated downregulation. Importantly, symptoms of preeclampsia were prevented by transgenic ARRB1 expression or a small-molecule drug. Because AT1-B2 heteromerization was found to occur in human placental biopsies from pregnancies complicated by preeclampsia, specifically targeting AT1-B2 heteromerization and its downstream consequences represents a promising therapeutic approach.


Subject(s)
Angiotensin II/metabolism , Receptor, Bradykinin B2/metabolism , beta-Arrestin 1/metabolism , Animals , Calcium Signaling , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Oligopeptides , Placenta/metabolism , Pre-Eclampsia/prevention & control , Pregnancy , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/physiology , beta-Arrestin 1/genetics , beta-Arrestin 1/physiology
5.
Cell ; 178(1): 202-215.e14, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31204102

ABSTRACT

Despite the worldwide success of vaccination, newborns remain vulnerable to infections. While neonatal vaccination has been hampered by maternal antibody-mediated dampening of immune responses, enhanced regulatory and tolerogenic mechanisms, and immune system immaturity, maternal pre-natal immunization aims to boost neonatal immunity via antibody transfer to the fetus. However, emerging data suggest that antibodies are not transferred equally across the placenta. To understand this, we used systems serology to define Fc features associated with antibody transfer. The Fc-profile of neonatal and maternal antibodies differed, skewed toward natural killer (NK) cell-activating antibodies. This selective transfer was linked to digalactosylated Fc-glycans that selectively bind FcRn and FCGR3A, resulting in transfer of antibodies able to efficiently leverage innate immune cells present at birth. Given emerging data that vaccination may direct antibody glycosylation, our study provides insights for the development of next-generation maternal vaccines designed to elicit antibodies that will most effectively aid neonates.


Subject(s)
Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Placenta/metabolism , Polysaccharides/metabolism , Receptors, Fc/immunology , Receptors, Fc/metabolism , Adolescent , Adult , Belgium , Cell Degranulation , Cohort Studies , Female , Glycosylation , Humans , Infant, Newborn , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Male , Pregnancy , Receptors, IgG/metabolism , THP-1 Cells , United States , Vaccination , Young Adult
6.
Cell ; 178(1): 190-201.e11, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31204101

ABSTRACT

The placental transfer of maternal IgG is critical for infant protection against infectious pathogens. However, factors that modulate the placental transfer of IgG remain largely undefined. HIV-infected women have impaired placental IgG transfer, presenting a unique "disruption model" to define factors that modulate placental IgG transfer. We measured the placental transfer efficiency of maternal HIV and pathogen-specific IgG in US and Malawian HIV-infected mothers and their HIV-exposed uninfected and infected infants. We examined the role of maternal HIV disease progression, infant factors, placental Fc receptor expression, IgG subclass, and glycan signatures and their association with placental IgG transfer efficiency. Maternal IgG characteristics, such as binding to placentally expressed Fc receptors FcγRIIa and FcγRIIIa, and Fc region glycan profiles were associated with placental IgG transfer efficiency. Our findings suggest that Fc region characteristics modulate the selective placental transfer of IgG, with implications for maternal vaccine design and infant health.


Subject(s)
HIV Infections/transmission , HIV/genetics , Immunoglobulin G/blood , Infectious Disease Transmission, Vertical , Placenta/metabolism , Pregnancy Complications, Infectious/virology , Receptors, IgG/metabolism , Cohort Studies , Disease Progression , Female , Glycosylation , HIV Infections/immunology , HIV Infections/virology , Humans , Immunoglobulin Fc Fragments/metabolism , Infant , Infant, Newborn , Malawi , Pregnancy , Pregnancy Complications, Infectious/immunology , United States , Viral Load/genetics
7.
Physiol Rev ; 103(4): 2523-2560, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37171808

ABSTRACT

The placenta is a unique organ system that functionally combines both maternal and fetal cell types with distinct lineage origins. Normal placentation is critical for developmental progression and reproductive success. Although the placenta is best known for its nutrient supply function to the fetus, genetic experiments in mice highlight that the placenta is also pivotal for directing the proper formation of specific fetal organs. These roles underscore the importance of the placenta for pregnancy outcome and lifelong health span, which makes it essential to better understand the molecular processes governing placental development and function and to find adequate models to study it. In this review, we provide an overview of placental development and highlight the instructional role of the epigenome in dictating cell fate decisions specifically in the placental trophoblast cell lineage. We then focus on recent advances in exploring stem cell and organoid models reflecting the feto-maternal interface in mice and humans that provide much-improved tools to study events in early development. We discuss stem cells derived from the placenta as well as those artificially induced to resemble the placenta, and how they can be combined with embryonic stem cells and with endometrial cell types of the uterus to reconstitute the early implantation site. We then allude to the exciting prospects of how these models can be harnessed in biomedicine to enhance our understanding of the pathological underpinnings of pregnancy complications in a patient-specific manner, and ultimately to facilitate therapeutic approaches of tissue- and organ-based regenerative medicine.


Subject(s)
Placenta , Trophoblasts , Pregnancy , Female , Humans , Animals , Mice , Placenta/metabolism , Trophoblasts/metabolism , Trophoblasts/pathology , Placentation , Cell Differentiation , Epigenesis, Genetic
8.
Nature ; 625(7996): 760-767, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38092039

ABSTRACT

GDF15, a hormone acting on the brainstem, has been implicated in the nausea and vomiting of pregnancy, including its most severe form, hyperemesis gravidarum (HG), but a full mechanistic understanding is lacking1-4. Here we report that fetal production of GDF15 and maternal sensitivity to it both contribute substantially to the risk of HG. We confirmed that higher GDF15 levels in maternal blood are associated with vomiting in pregnancy and HG. Using mass spectrometry to detect a naturally labelled GDF15 variant, we demonstrate that the vast majority of GDF15 in the maternal plasma is derived from the feto-placental unit. By studying carriers of rare and common genetic variants, we found that low levels of GDF15 in the non-pregnant state increase the risk of developing HG. Conversely, women with ß-thalassaemia, a condition in which GDF15 levels are chronically high5, report very low levels of nausea and vomiting of pregnancy. In mice, the acute food intake response to a bolus of GDF15 is influenced bi-directionally by prior levels of circulating GDF15 in a manner suggesting that this system is susceptible to desensitization. Our findings support a putative causal role for fetally derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by prepregnancy exposure to the hormone, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.


Subject(s)
Growth Differentiation Factor 15 , Hyperemesis Gravidarum , Nausea , Vomiting , Animals , Female , Humans , Mice , Pregnancy , beta-Thalassemia/blood , beta-Thalassemia/metabolism , Fetus/metabolism , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/metabolism , Hormones/blood , Hormones/metabolism , Hyperemesis Gravidarum/complications , Hyperemesis Gravidarum/metabolism , Hyperemesis Gravidarum/prevention & control , Hyperemesis Gravidarum/therapy , Nausea/blood , Nausea/complications , Nausea/metabolism , Placenta/metabolism , Vomiting/blood , Vomiting/complications , Vomiting/metabolism
9.
Nature ; 629(8012): 652-659, 2024 May.
Article in English | MEDLINE | ID: mdl-38693261

ABSTRACT

The gut microbiota operates at the interface of host-environment interactions to influence human homoeostasis and metabolic networks1-4. Environmental factors that unbalance gut microbial ecosystems can therefore shape physiological and disease-associated responses across somatic tissues5-9. However, the systemic impact of the gut microbiome on the germline-and consequently on the F1 offspring it gives rise to-is unexplored10. Here we show that the gut microbiota act as a key interface between paternal preconception environment and intergenerational health in mice. Perturbations to the gut microbiota of prospective fathers increase the probability of their offspring presenting with low birth weight, severe growth restriction and premature mortality. Transmission of disease risk occurs via the germline and is provoked by pervasive gut microbiome perturbations, including non-absorbable antibiotics or osmotic laxatives, but is rescued by restoring the paternal microbiota before conception. This effect is linked with a dynamic response to induced dysbiosis in the male reproductive system, including impaired leptin signalling, altered testicular metabolite profiles and remapped small RNA payloads in sperm. As a result, dysbiotic fathers trigger an elevated risk of in utero placental insufficiency, revealing a placental origin of mammalian intergenerational effects. Our study defines a regulatory 'gut-germline axis' in males, which is sensitive to environmental exposures and programmes offspring fitness through impacting placenta function.


Subject(s)
Disease Susceptibility , Dysbiosis , Fathers , Gastrointestinal Microbiome , Placental Insufficiency , Prenatal Injuries , Spermatozoa , Animals , Female , Male , Mice , Pregnancy , Dysbiosis/complications , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Leptin/metabolism , Mice, Inbred C57BL , Placenta/metabolism , Placenta/physiopathology , Placental Insufficiency/etiology , Placental Insufficiency/metabolism , Placental Insufficiency/physiopathology , Pregnancy Outcome , Prenatal Injuries/etiology , Prenatal Injuries/metabolism , Prenatal Injuries/physiopathology , Signal Transduction , Spermatozoa/metabolism , Testis/metabolism , Testis/physiopathology , Disease Susceptibility/etiology
10.
Physiol Rev ; 101(3): 739-795, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33270534

ABSTRACT

Almost 2 billion adults in the world are overweight, and more than half of them are classified as obese, while nearly one-third of children globally experience poor growth and development. Given the vast amount of knowledge that has been gleaned from decades of research on growth and development, a number of questions remain as to why the world is now in the midst of a global epidemic of obesity accompanied by the "double burden of malnutrition," where overweight coexists with underweight and micronutrient deficiencies. This challenge to the human condition can be attributed to nutritional and environmental exposures during pregnancy that may program a fetus to have a higher risk of chronic diseases in adulthood. To explore this concept, frequently called the developmental origins of health and disease (DOHaD), this review considers a host of factors and physiological mechanisms that drive a fetus or child toward a higher risk of obesity, fatty liver disease, hypertension, and/or type 2 diabetes (T2D). To that end, this review explores the epidemiology of DOHaD with discussions focused on adaptations to human energetics, placental development, dysmetabolism, and key environmental exposures that act to promote chronic diseases in adulthood. These areas are complementary and additive in understanding how providing the best conditions for optimal growth can create the best possible conditions for lifelong health. Moreover, understanding both physiological as well as epigenetic and molecular mechanisms for DOHaD is vital to most fully address the global issues of obesity and other chronic diseases.


Subject(s)
Metabolic Diseases/etiology , Obesity/etiology , Placenta/metabolism , Female , Humans , Metabolic Diseases/metabolism , Obesity/metabolism , Pregnancy
11.
Nature ; 604(7905): 349-353, 2022 04.
Article in English | MEDLINE | ID: mdl-35388219

ABSTRACT

Mammalian embryogenesis requires rapid growth and proper metabolic regulation1. Midgestation features increasing oxygen and nutrient availability concomitant with fetal organ development2,3. Understanding how metabolism supports development requires approaches to observe metabolism directly in model organisms in utero. Here we used isotope tracing and metabolomics to identify evolving metabolic programmes in the placenta and embryo during midgestation in mice. These tissues differ metabolically throughout midgestation, but we pinpointed gestational days (GD) 10.5-11.5 as a transition period for both placenta and embryo. Isotope tracing revealed differences in carbohydrate metabolism between the tissues and rapid glucose-dependent purine synthesis, especially in the embryo. Glucose's contribution to the tricarboxylic acid (TCA) cycle rises throughout midgestation in the embryo but not in the placenta. By GD12.5, compartmentalized metabolic programmes are apparent within the embryo, including different nutrient contributions to the TCA cycle in different organs. To contextualize developmental anomalies associated with Mendelian metabolic defects, we analysed mice deficient in LIPT1, the enzyme that activates 2-ketoacid dehydrogenases related to the TCA cycle4,5. LIPT1 deficiency suppresses TCA cycle metabolism during the GD10.5-GD11.5 transition, perturbs brain, heart and erythrocyte development and leads to embryonic demise by GD11.5. These data document individualized metabolic programmes in developing organs in utero.


Subject(s)
Citric Acid Cycle , Fetal Development , Metabolomics , Placenta , Animals , Embryo, Mammalian/metabolism , Female , Glucose/metabolism , Mammals/metabolism , Mice , Placenta/metabolism , Pregnancy
12.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38112206

ABSTRACT

Placental development involves coordinated expansion and differentiation of trophoblast cell lineages possessing specialized functions. Among the differentiated trophoblast cell lineages are invasive trophoblast cells, which exit the placenta and invade the uterus, where they restructure the uterine parenchyma and facilitate remodeling of uterine spiral arteries. The rat exhibits deep intrauterine trophoblast cell invasion, a feature shared with human placentation, and is also amenable to gene manipulation using genome-editing techniques. In this investigation, we generated a conditional rat model targeting the invasive trophoblast cell lineage. Prolactin family 7, subfamily b, member 1 (Prl7b1) is uniquely and abundantly expressed in the rat invasive trophoblast cell lineage. Disruption of Prl7b1 did not adversely affect placental development. We demonstrated that the Prl7b1 locus could be effectively used to drive the expression of Cre recombinase in invasive trophoblast cells. Our rat model represents a new tool for investigating candidate genes contributing to the regulation of invasive trophoblast cells and their roles in trophoblast-guided uterine spiral artery remodeling.


Subject(s)
Placenta , Placentation , Pregnancy , Rats , Female , Animals , Humans , Placenta/metabolism , Placentation/genetics , Trophoblasts , Uterus , Cell Lineage/genetics , Models, Animal
13.
Development ; 151(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38300826

ABSTRACT

ACKR3 scavenges and degrades the stem cell recruiting chemokine CXCL12, which is essential for proper embryonic and, in particular, haematopoietic development. Here, we demonstrate strong expression of ACKR3 on trophoblasts. Using a maternally administered pharmacological blocker and Cre-mediated genetic approaches, we demonstrate that trophoblast ACKR3 is essential for preventing movement of CXCL12 from the mother to the embryo, with elevated plasma CXCL12 levels being detected in embryos from ACKR3-blocker-treated mothers. Mice born to mothers treated with the blocker are lighter and shorter than those born to vehicle-treated mothers and, in addition, display profound anaemia associated with a markedly reduced bone marrow haematopoietic stem cell population. Importantly, although the haematopoietic abnormalities are corrected as mice age, our studies reveal a postnatal window during which offspring of ACKR3-blocker-treated mice are unable to mount effective inflammatory responses to inflammatory/infectious stimuli. Overall, these data demonstrate that ACKR3 is essential for preventing CXCL12 transfer from mother to embryo and for ensuring properly regulated CXCL12 control over the development of the haematopoietic system.


Subject(s)
Placenta , Receptors, CXCR , Animals , Female , Mice , Pregnancy , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Movement , Mutation , Placenta/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Signal Transduction/genetics
14.
PLoS Biol ; 22(1): e3002464, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38206904

ABSTRACT

Trichromacy is unique to primates among placental mammals, enabled by blue (short/S), green (medium/M), and red (long/L) cones. In humans, great apes, and Old World monkeys, cones make a poorly understood choice between M and L cone subtype fates. To determine mechanisms specifying M and L cones, we developed an approach to visualize expression of the highly similar M- and L-opsin mRNAs. M-opsin was observed before L-opsin expression during early human eye development, suggesting that M cones are generated before L cones. In adult human tissue, the early-developing central retina contained a mix of M and L cones compared to the late-developing peripheral region, which contained a high proportion of L cones. Retinoic acid (RA)-synthesizing enzymes are highly expressed early in retinal development. High RA signaling early was sufficient to promote M cone fate and suppress L cone fate in retinal organoids. Across a human population sample, natural variation in the ratios of M and L cone subtypes was associated with a noncoding polymorphism in the NR2F2 gene, a mediator of RA signaling. Our data suggest that RA promotes M cone fate early in development to generate the pattern of M and L cones across the human retina.


Subject(s)
Placenta , Tretinoin , Pregnancy , Adult , Animals , Humans , Female , Tretinoin/metabolism , Placenta/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retina/metabolism , Opsins/metabolism , Rod Opsins/genetics , Primates , Mammals/metabolism
15.
Nature ; 592(7852): 80-85, 2021 04.
Article in English | MEDLINE | ID: mdl-33692543

ABSTRACT

Placentas can exhibit chromosomal aberrations that are absent from the fetus1. The basis of this genetic segregation, which is known as confined placental mosaicism, remains unknown. Here we investigated the phylogeny of human placental cells as reconstructed from somatic mutations, using whole-genome sequencing of 86 bulk placental samples (with a median weight of 28 mg) and of 106 microdissections of placental tissue. We found that every bulk placental sample represents a clonal expansion that is genetically distinct, and exhibits a genomic landscape akin to that of childhood cancer in terms of mutation burden and mutational imprints. To our knowledge, unlike any other healthy human tissue studied so far, the placental genomes often contained changes in copy number. We reconstructed phylogenetic relationships between tissues from the same pregnancy, which revealed that developmental bottlenecks genetically isolate placental tissues by separating trophectodermal lineages from lineages derived from the inner cell mass. Notably, there were some cases with full segregation-within a few cell divisions of the zygote-of placental lineages and lineages derived from the inner cell mass. Such early embryonic bottlenecks may enable the normalization of zygotic aneuploidy. We observed direct evidence for this in a case of mosaic trisomic rescue. Our findings reveal extensive mutagenesis in placental tissues and suggest that mosaicism is a typical feature of placental development.


Subject(s)
Mosaicism , Mutagenesis , Mutation , Placenta/metabolism , Biopsy , Blastocyst Inner Cell Mass/cytology , Female , Genome, Human/genetics , Humans , Mesoderm/cytology , Mutation Rate , Placenta/cytology , Pregnancy , Trisomy/genetics , Trophoblasts/cytology , Trophoblasts/metabolism , Zygote/cytology
16.
Proc Natl Acad Sci U S A ; 121(28): e2404062121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968109

ABSTRACT

Nutrient sensing and adaptation in the placenta are essential for pregnancy viability and proper fetal growth. Our recent study demonstrated that the placenta adapts to nutrient insufficiency through mechanistic target of rapamycin (mTOR) inhibition-mediated trophoblast differentiation toward syncytiotrophoblasts (STBs), a highly specialized multinucleated trophoblast subtype mediating extensive maternal-fetal interactions. However, the underlying mechanism remains elusive. Here, we unravel the indispensable role of the mTORC1 downstream transcriptional factor TFEB in STB formation both in vitro and in vivo. TFEB deficiency significantly impaired STB differentiation in human trophoblasts and placenta organoids. Consistently, systemic or trophoblast-specific deletion of Tfeb compromised STB formation and placental vascular construction, leading to severe embryonic lethality. Mechanistically, TFEB conferred direct transcriptional activation of the fusogen ERVFRD-1 in human trophoblasts and thereby promoted STB formation, independent of its canonical function as a master regulator of the autophagy-lysosomal pathway. Moreover, we demonstrated that TFEB directed the trophoblast syncytialization response driven by mTOR complex 1 (mTORC1) signaling. TFEB expression positively correlated with the reinforced trophoblast syncytialization in human fetal growth-restricted placentas exhibiting suppressed mTORC1 activity. Our findings substantiate that the TFEB-fusogen axis ensures proper STB formation during placenta development and under nutrient stress, shedding light on TFEB as a mechanistic link between nutrient-sensing machinery and trophoblast differentiation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Differentiation , Mechanistic Target of Rapamycin Complex 1 , Trophoblasts , Trophoblasts/metabolism , Humans , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Female , Pregnancy , Mice , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Placenta/metabolism , Signal Transduction , Autophagy/physiology
17.
Proc Natl Acad Sci U S A ; 121(12): e2318176121, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38483994

ABSTRACT

Endogenous retroviruses (ERVs) are frequently reactivated in mammalian placenta. It has been proposed that ERVs contribute to shaping the gene regulatory network of mammalian trophoblasts, dominantly acting as species- and placental-specific enhancers. However, whether and how ERVs control human trophoblast development through alternative pathways remains poorly understood. Besides the well-recognized function of human endogenous retrovirus-H (HERVH) in maintaining pluripotency of early human epiblast, here we present a unique role of HERVH on trophoblast lineage development. We found that the LTR7C/HERVH subfamily exhibits an accessible chromatin state in the human trophoblast lineage. Particularly, the LTR7C/HERVH-derived Urothelial Cancer Associated 1 (UCA1), a primate-specific long non-coding RNA (lncRNA), is transcribed in human trophoblasts and promotes the proliferation of human trophoblast stem cells (hTSCs), whereas its ectopic expression compromises human trophoblast syncytialization coinciding with increased interferon signaling pathway. Importantly, UCA1 upregulation is detectable in placental samples from early-onset preeclampsia (EO-PE) patients and the transcriptome of EO-PE placenta exhibits considerable similarities to that of the syncytiotrophoblasts differentiated from UCA1-overexpressing hTSCs, supporting up-regulated UCA1 as a potential biomarker of this disease. Altogether, our data shed light on the versatile regulatory role of HERVH in early human development and provide a unique mechanism whereby ERVs exert a function in human placentation and placental syndromes.


Subject(s)
Endogenous Retroviruses , RNA, Long Noncoding , Animals , Humans , Pregnancy , Female , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Placenta/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Trophoblasts/metabolism , Placentation , Primates/genetics , Mammals/genetics
18.
Development ; 150(11)2023 06 01.
Article in English | MEDLINE | ID: mdl-37278344

ABSTRACT

The placenta is essential for reproductive success. The murine placenta includes polyploid giant cells that are crucial for its function. Polyploidy occurs broadly in nature but its regulators and significance in the placenta are unknown. We have discovered that many murine placental cell types are polyploid and have identified factors that license polyploidy using single-cell RNA sequencing. Myc is a key regulator of polyploidy and placental development, and is required for multiple rounds of DNA replication, likely via endocycles, in trophoblast giant cells. Furthermore, MYC supports the expression of DNA replication and nucleotide biosynthesis genes along with ribosomal RNA. Increased DNA damage and senescence occur in trophoblast giant cells without Myc, accompanied by senescence in the neighboring maternal decidua. These data reveal Myc is essential for polyploidy to support normal placental development, thereby preventing premature senescence. Our study, combined with available literature, suggests that Myc is an evolutionarily conserved regulator of polyploidy.


Subject(s)
Placenta , Trophoblasts , Animals , Female , Mice , Pregnancy , Placenta/metabolism , Placentation , Polyploidy , Trophoblasts/metabolism
19.
Development ; 150(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37831056

ABSTRACT

Compelling epidemiological and animal experimental data demonstrate that cardiometabolic and neuropsychiatric diseases originate in a suboptimal intrauterine environment. Here, we review evidence suggesting that altered placental function may, at least in part, mediate the link between the maternal environment and changes in fetal growth and development. Emerging evidence indicates that the placenta controls the development and function of several fetal tissues through nutrient sensing, modulation of trophoblast nutrient transporters and by altering the number and cargo of released extracellular vesicles. In this Review, we discuss the development and functions of the maternal-placental-fetal interface (in humans and mice) and how cross-talk between these compartments may be a mechanism for in utero programming, focusing on mechanistic target of rapamycin (mTOR), adiponectin and O-GlcNac transferase (OGT) signaling. We also discuss how maternal diet and stress influences fetal development and metabolism and how fetal growth restriction can result in susceptibility to developing chronic disease later in life. Finally, we speculate how interventions targeting placental function may offer unprecedented opportunities to prevent cardiometabolic disease in future generations.


Subject(s)
Fetal Development , Placenta , Pregnancy , Female , Humans , Mice , Animals , Placenta/metabolism , Trophoblasts/metabolism , Signal Transduction , Fetal Growth Retardation
20.
Development ; 150(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37905445

ABSTRACT

Failures in growth and differentiation of the early human placenta are associated with severe pregnancy disorders such as pre-eclampsia and fetal growth restriction. However, regulatory mechanisms controlling development of placental epithelial cells, the trophoblasts, remain poorly elucidated. Using trophoblast stem cells (TSCs), trophoblast organoids (TB-ORGs) and primary cytotrophoblasts (CTBs) of early pregnancy, we herein show that autocrine NOTCH3 signalling controls human placental expansion and differentiation. The NOTCH3 receptor was specifically expressed in proliferative CTB progenitors and its active form, the nuclear NOTCH3 intracellular domain (NOTCH3-ICD), interacted with the transcriptional co-activator mastermind-like 1 (MAML1). Doxycycline-inducible expression of dominant-negative MAML1 in TSC lines provoked cell fusion and upregulation of genes specific for multinucleated syncytiotrophoblasts, which are the differentiated hormone-producing cells of the placenta. However, progenitor expansion and markers of trophoblast stemness and proliferation were suppressed. Accordingly, inhibition of NOTCH3 signalling diminished growth of TB-ORGs, whereas overexpression of NOTCH3-ICD in primary CTBs and TSCs showed opposite effects. In conclusion, the data suggest that canonical NOTCH3 signalling plays a key role in human placental development by promoting self-renewal of CTB progenitors.


Subject(s)
Placenta , Trophoblasts , Humans , Pregnancy , Female , Placenta/metabolism , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Stem Cells , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL