Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Publication year range
1.
Cell ; 165(4): 1012-26, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27062923

ABSTRACT

Mouse studies have been instrumental in forming our current understanding of early cell-lineage decisions; however, similar insights into the early human development are severely limited. Here, we present a comprehensive transcriptional map of human embryo development, including the sequenced transcriptomes of 1,529 individual cells from 88 human preimplantation embryos. These data show that cells undergo an intermediate state of co-expression of lineage-specific genes, followed by a concurrent establishment of the trophectoderm, epiblast, and primitive endoderm lineages, which coincide with blastocyst formation. Female cells of all three lineages achieve dosage compensation of X chromosome RNA levels prior to implantation. However, in contrast to the mouse, XIST is transcribed from both alleles throughout the progression of this expression dampening, and X chromosome genes maintain biallelic expression while dosage compensation proceeds. We envision broad utility of this transcriptional atlas in future studies on human development as well as in stem cell research.


Subject(s)
Blastocyst/metabolism , Chromosomes, Human, X , Single-Cell Analysis , Blastocyst Inner Cell Mass/metabolism , Dosage Compensation, Genetic , Female , Humans , Male , RNA, Long Noncoding/genetics , Sequence Analysis, RNA , Sex Characteristics , Transcriptome
2.
EMBO Rep ; 25(4): 1752-1772, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38491313

ABSTRACT

Emerging evidence indicates that parental diseases can impact the health of subsequent generations through epigenetic inheritance. Recently, it was shown that maternal diabetes alters the metaphase II oocyte transcriptome, causing metabolic dysfunction in offspring. However, type 1 diabetes (T1D) mouse models frequently utilized in previous studies may be subject to several confounding factors due to severe hyperglycemia. This limits clinical translatability given improvements in glycemic control for T1D subjects. Here, we optimize a T1D mouse model to investigate the effects of appropriately managed maternal glycemic levels on oocytes and intrauterine development. We show that diabetic mice with appropriate glycemic control exhibit better long-term health, including maintenance of the oocyte transcriptome and chromatin accessibility. We further show that human oocytes undergoing in vitro maturation challenged with mildly increased levels of glucose, reflecting appropriate glycemic management, also retain their transcriptome. However, fetal growth and placental function are affected in mice despite appropriate glycemic control, suggesting the uterine environment rather than the germline as a pathological factor in developmental programming in appropriately managed diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Hyperglycemia , Humans , Female , Pregnancy , Mice , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Placenta , Hyperglycemia/genetics , Hyperglycemia/metabolism , Oocytes/metabolism , Disease Models, Animal
4.
BMC Biol ; 21(1): 277, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38031019

ABSTRACT

BACKGROUND: Estrogen receptor beta (ERß, Esr2) plays a pivotal role in folliculogenesis and ovulation, yet its exact mechanism of action is mainly uncharacterized. RESULTS: We here performed ERß ChIP-sequencing of mouse ovaries followed by complementary RNA-sequencing of wild-type and ERß knockout ovaries. By integrating the ERß cistrome and transcriptome, we identified its direct target genes and enriched biological functions in the ovary. This demonstrated its strong impact on genes regulating organism development, cell migration, lipid metabolism, response to hypoxia, and response to estrogen. Cell-type deconvolution analysis of the bulk RNA-seq data revealed a decrease in luteal cells and an increased proportion of theca cells and a specific type of cumulus cells upon ERß loss. Moreover, we identified a significant overlap with the gene regulatory network of liver receptor homolog 1 (LRH-1, Nr5a2) and showed that ERß and LRH-1 extensively bound to the same chromatin locations in granulosa cells. Using ChIP-reChIP, we corroborated simultaneous ERß and LRH-1 co-binding at the ERß-repressed gene Greb1 but not at the ERß-upregulated genes Cyp11a1 and Fkbp5. Transactivation assay experimentation further showed that ERß and LRH-1 can inhibit their respective transcriptional activity at classical response elements. CONCLUSIONS: By characterizing the genome-wide endogenous ERß chromatin binding, gene regulations, and extensive crosstalk between ERß and LRH-1, along with experimental corroborations, our data offer genome-wide mechanistic underpinnings of ovarian physiology and fertility.


Subject(s)
Estrogen Receptor beta , Ovary , Animals , Female , Mice , Chromatin/genetics , Estrogen Receptor beta/genetics , Gene Expression Regulation , Transcriptome
5.
Genome Res ; 30(8): 1083-1096, 2020 08.
Article in English | MEDLINE | ID: mdl-32820007

ABSTRACT

Somatic motor neurons are selectively vulnerable in spinal muscular atrophy (SMA), which is caused by a deficiency of the ubiquitously expressed survival of motor neuron protein. However, some motor neuron groups, including oculomotor and trochlear (ocular), which innervate eye muscles, are for unknown reasons spared. To reveal mechanisms of vulnerability and resistance in SMA, we investigate the transcriptional dynamics in discrete neuronal populations using laser capture microdissection coupled with RNA sequencing (LCM-seq). Using gene correlation network analysis, we reveal a TRP53-mediated stress response that is intrinsic to all somatic motor neurons independent of their vulnerability, but absent in relatively resistant red nucleus and visceral motor neurons. However, the temporal and spatial expression analysis across neuron types shows that the majority of SMA-induced modulations are cell type-specific. Using Gene Ontology and protein network analyses, we show that ocular motor neurons present unique disease-adaptation mechanisms that could explain their resilience. Specifically, ocular motor neurons up-regulate (1) Syt1, Syt5, and Cplx2, which modulate neurotransmitter release; (2) the neuronal survival factors Gdf15, Chl1, and Lif; (3) Aldh4, that protects cells from oxidative stress; and (4) the caspase inhibitor Pak4. Finally, we show that GDF15 can rescue vulnerable human spinal motor neurons from degeneration. This confirms that adaptation mechanisms identified in resilient neurons can be used to reduce susceptibility of vulnerable neurons. In conclusion, this in-depth longitudinal transcriptomics analysis in SMA reveals novel cell type-specific changes that, alone and combined, present compelling targets, including Gdf15, for future gene therapy studies aimed toward preserving vulnerable motor neurons.


Subject(s)
Adaptation, Physiological/physiology , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Neuroprotection/genetics , Adaptation, Physiological/genetics , Animals , Cells, Cultured , Disease Models, Animal , Eye/innervation , Genetic Predisposition to Disease/genetics , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Laser Capture Microdissection , Mice , Mice, Knockout , Motor Cortex/pathology , Sequence Analysis, RNA , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Diabetologia ; 65(9): 1483-1494, 2022 09.
Article in English | MEDLINE | ID: mdl-35771237

ABSTRACT

AIMS/HYPOTHESIS: The link underlying abnormal glucose metabolism, type 2 diabetes and polycystic ovary syndrome (PCOS) that is independent of BMI remains unclear in observational studies. We aimed to clarify this association using a genome-wide cross-trait approach. METHODS: Summary statistics from the hitherto largest genome-wide association studies conducted for type 2 diabetes, type 2 diabetes mellitus adjusted for BMI (T2DMadjBMI), fasting glucose, fasting insulin, 2h glucose after an oral glucose challenge (all adjusted for BMI), HbA1c and PCOS, all in populations of European ancestry, were used. We quantified overall and local genetic correlations, identified pleiotropic loci and expression-trait associations, and made causal inferences across traits. RESULTS: A positive overall genetic correlation between type 2 diabetes and PCOS was observed, largely influenced by BMI (rg=0.31, p=1.63×10-8) but also independent of BMI (T2DMadjBMI-PCOS: rg=0.12, p=0.03). Sixteen pleiotropic loci affecting type 2 diabetes, glycaemic traits and PCOS were identified, suggesting mechanisms of association that are independent of BMI. Two shared expression-trait associations were found for type 2 diabetes/T2DMadjBMI and PCOS targeting tissues of the cardiovascular, exocrine/endocrine and digestive systems. A putative causal effect of fasting insulin adjusted for BMI and type 2 diabetes on PCOS was demonstrated. CONCLUSIONS/INTERPRETATION: We found a genetic link underlying type 2 diabetes, glycaemic traits and PCOS, driven by both biological pleiotropy and causal mediation, some of which is independent of BMI. Our findings highlight the importance of controlling fasting insulin levels to mitigate the risk of PCOS, as well as screening for and long-term monitoring of type 2 diabetes in all women with PCOS, irrespective of BMI.


Subject(s)
Diabetes Mellitus, Type 2 , Polycystic Ovary Syndrome , Blood Glucose , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Genome-Wide Association Study , Humans , Insulin/genetics , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism
7.
BMC Med ; 20(1): 66, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35144605

ABSTRACT

BACKGROUND: The comorbidity between polycystic ovary syndrome (PCOS) and obesity has long been observed in clinical settings, but their shared genetic basis remains unclear. METHODS: Leveraging summary statistics of large-scale GWAS(s) conducted in European-ancestry populations on body mass index (adult BMI, Nfemale=434,794; childhood BMI, N=39,620), waist-to-hip ratio (WHR, Nfemale=381,152), WHR adjusted for BMI (WHRadjBMI, Nfemale=379,501), and PCOS (Ncase=10,074, Ncontrol=103,164), we performed a large-scale genome-wide cross-trait analysis to quantify overall and local genetic correlation, to identify shared loci, and to infer causal relationship. RESULTS: We found positive genetic correlations between PCOS and adult BMI (rg=0.47, P=2.19×10-16), childhood BMI (rg=0.31, P=6.72×10-5), and WHR (rg=0.32, P=1.34×10-10), all withstanding Bonferroni correction. A suggestive significant genetic correlation was found between PCOS and WHRadjBMI (rg=0.09, P=0.04). Partitioning the whole genome into 1703 nearly independent regions, we observed a significant local genetic correlation for adult BMI and PCOS at chromosome 18: 57630483-59020751. We identified 16 shared loci underlying PCOS and obesity-related traits via cross-trait meta-analysis including 9 loci shared between BMI and PCOS (adult BMI and PCOS: 5 loci; childhood BMI and PCOS: 4 loci), 6 loci shared between WHR and PCOS, and 5 loci shared between WHRadjBMI and PCOS. Mendelian randomization (MR) supported the causal roles of both adult BMI (OR=2.92, 95% CI=2.33-3.67) and childhood BMI (OR=2.76, 95% CI=2.09-3.66) in PCOS, but not WHR (OR=1.19, 95% CI=0.93-1.52) or WHRadjBMI (OR=1.03, 95% CI=0.87-1.22). Genetic predisposition to PCOS did not seem to influence the risk of obesity-related traits. CONCLUSIONS: Our cross-trait analysis suggests a shared genetic basis underlying obesity and PCOS and provides novel insights into the biological mechanisms underlying these complex traits. Our work informs public health intervention by confirming the important role of weight management in PCOS prevention.


Subject(s)
Polycystic Ovary Syndrome , Adult , Body Mass Index , Child , Female , Genome-Wide Association Study , Genomics , Humans , Obesity/epidemiology , Obesity/genetics , Polycystic Ovary Syndrome/epidemiology , Polycystic Ovary Syndrome/genetics , Polymorphism, Single Nucleotide/genetics
8.
FASEB J ; 34(11): 14440-14457, 2020 11.
Article in English | MEDLINE | ID: mdl-32892421

ABSTRACT

Nerve growth factor (NGF) is critical for the development and maintenance of the peripheral sympathetic neurons. NGF is also involved in the ovarian sympathetic innervation and in the development and maintenance of folliculogenesis. Women with the endocrine disorder, polycystic ovary syndrome (PCOS), have an increased sympathetic nerve activity and increased ovarian NGF levels. The role of ovarian NGF excess in the PCOS pathophysiology and in the PCOS-related features is unclear. Here, using transgenic mice overexpressesing NGF in the ovarian theca cells (17NF mice), we assessed the female embryonic development, and the reproductive and metabolic profile in adult females. Ovarian NGF excess caused growth restriction in the female fetuses, and a delayed gonocyte and primary oocyte maturation. In adulthood, the 17NF mice displayed irregular estrous cycles and altered ovarian expression of steroidogenic and epigenetic markers. They also exhibited an increased sympathetic output with increased circulating dopamine, and metabolic dysfunction reflected by aberrant adipose tissue morphology and function, impaired glucose metabolism, decreased energy expenditure, and hepatic steatosis. These findings indicate that ovarian NGF excess leads to adverse fetal development and to reproductive and metabolic complications in adulthood, mirroring common features of PCOS. This work provides evidence that NGF excess may be implicated in the PCOS pathophysiology.


Subject(s)
Fetal Development , Nerve Growth Factor/genetics , Ovary/metabolism , Polycystic Ovary Syndrome/genetics , Animals , Cells, Cultured , Dopamine/metabolism , Estrous Cycle , Female , Mice , Nerve Growth Factor/metabolism , Oogenesis , Ovary/embryology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Up-Regulation
9.
Bioorg Med Chem Lett ; 32: 127668, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33161125

ABSTRACT

Cholesteryl ester transfer protein (CETP) inhibitors reduce the transfer of cholesteryl esters from the high-density lipoprotein (HDL-C) to apolipoprotein such as VLDL/LDL, with exchange of triglycerides. Thus, this inhibition increases the HDL-C levels, which is believed to lower the risk for heart disease and stroke. We report here a series of CETP inhibitors based on the cyclic, bicyclic urea and sulfamide cores. These CETP inhibitors exemplified by 15, 31, and 45 demonstrated in vitro potency in inhibiting the CETP transfer activity, and 15, 31 showing in vivo efficacy to increase HDL-C levels in cynomolgus-CETP transgenic mice. The synthesis and biological evaluations of these CETP inhibitors are described.


Subject(s)
Anticholesteremic Agents/chemical synthesis , Bridged Bicyclo Compounds/chemistry , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Sulfonamides/chemistry , Urea/analogs & derivatives , Animals , Anticholesteremic Agents/metabolism , Anticholesteremic Agents/therapeutic use , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol, HDL/blood , Cyclization , Dyslipidemias/drug therapy , Dyslipidemias/pathology , Humans , Mice , Mice, Transgenic , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/therapeutic use , Urea/metabolism , Urea/therapeutic use
10.
Bioorg Med Chem Lett ; 42: 128046, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33865969

ABSTRACT

PI3K-δ mediates key immune cell signaling pathways and is a target of interest for treatment of oncological and immunological disorders. Here we describe the discovery and optimization of a novel series of PI3K-δ selective inhibitors. We first identified hits containing an isoindolinone scaffold using a combined ligand- and receptor-based virtual screening workflow, and then improved potency and selectivity guided by structural data and modeling. Careful optimization of molecular properties led to compounds with improved permeability and pharmacokinetic profile, and high potency in a whole blood assay.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Discovery , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phthalimides/pharmacology , Class I Phosphatidylinositol 3-Kinases/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/chemistry , Phthalimides/chemical synthesis , Phthalimides/chemistry , Structure-Activity Relationship
11.
Arterioscler Thromb Vasc Biol ; 39(7): 1402-1418, 2019 07.
Article in English | MEDLINE | ID: mdl-31242036

ABSTRACT

Objective- Pathological neovascularization is crucial for progression and morbidity of serious diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. While mechanisms of ongoing pathological neovascularization have been extensively studied, the initiating pathological vascular remodeling (PVR) events, which precede neovascularization remains poorly understood. Here, we identify novel molecular and cellular mechanisms of preneovascular PVR, by using the adult choriocapillaris as a model. Approach and Results- Using hypoxia or forced overexpression of VEGF (vascular endothelial growth factor) in the subretinal space to induce PVR in zebrafish and rats respectively, and by analyzing choriocapillaris membranes adjacent to choroidal neovascular lesions from age-related macular degeneration patients, we show that the choriocapillaris undergo robust induction of vascular intussusception and permeability at preneovascular stages of PVR. This PVR response included endothelial cell proliferation, formation of endothelial luminal processes, extensive vesiculation and thickening of the endothelium, degradation of collagen fibers, and splitting of existing extravascular columns. RNA-sequencing established a role for endothelial tight junction disruption, cytoskeletal remodeling, vesicle- and cilium biogenesis in this process. Mechanistically, using genetic gain- and loss-of-function zebrafish models and analysis of primary human choriocapillaris endothelial cells, we determined that HIF (hypoxia-induced factor)-1α-VEGF-A-VEGFR2 signaling was important for hypoxia-induced PVR. Conclusions- Our findings reveal that PVR involving intussusception and splitting of extravascular columns, endothelial proliferation, vesiculation, fenestration, and thickening is induced before neovascularization, suggesting that identifying and targeting these processes may prevent development of advanced neovascular disease in the future. Visual Overview- An online visual overview is available for this article.


Subject(s)
Neovascularization, Pathologic/etiology , Vascular Remodeling/physiology , Adult , Animals , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Macular Degeneration/etiology , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , Zebrafish
12.
Genome Res ; 26(10): 1342-1354, 2016 10.
Article in English | MEDLINE | ID: mdl-27486082

ABSTRACT

Pluripotency, differentiation, and X Chromosome inactivation (XCI) are key aspects of embryonic development. However, the underlying relationship and mechanisms among these processes remain unclear. Here, we systematically dissected these features along developmental progression using mouse embryonic stem cells (mESCs) and single-cell RNA sequencing with allelic resolution. We found that mESCs grown in a ground state 2i condition displayed transcriptomic profiles diffused from preimplantation mouse embryonic cells, whereas EpiStem cells closely resembled the post-implantation epiblast. Sex-related gene expression varied greatly across distinct developmental states. We also identified novel markers that were highly enriched in each developmental state. Moreover, we revealed that several novel pathways, including PluriNetWork and Focal Adhesion, were responsible for the delayed progression of female EpiStem cells. Importantly, we "digitalized" XCI progression using allelic expression of active and inactive X Chromosomes and surprisingly found that XCI states exhibited profound variability in each developmental state, including the 2i condition. XCI progression was not tightly synchronized with loss of pluripotency and increase of differentiation at the single-cell level, although these processes were globally correlated. In addition, highly expressed genes, including core pluripotency factors, were in general biallelically expressed. Taken together, our study sheds light on the dynamics of XCI progression and the asynchronicity between pluripotency, differentiation, and XCI.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , X Chromosome Inactivation , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells/metabolism , Single-Cell Analysis , Transcriptome
13.
Proc Natl Acad Sci U S A ; 113(30): E4387-96, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27407143

ABSTRACT

The LIM-homeodomain transcription factors Lmx1a and Lmx1b play critical roles during the development of midbrain dopaminergic progenitors, but their functions in the adult brain remain poorly understood. We show here that sustained expression of Lmx1a and Lmx1b is required for the survival of adult midbrain dopaminergic neurons. Strikingly, inactivation of Lmx1a and Lmx1b recreates cellular features observed in Parkinson's disease. We found that Lmx1a/b control the expression of key genes involved in mitochondrial functions, and their ablation results in impaired respiratory chain activity, increased oxidative stress, and mitochondrial DNA damage. Lmx1a/b deficiency caused axonal pathology characterized by α-synuclein(+) inclusions, followed by a progressive loss of dopaminergic neurons. These results reveal the key role of these transcription factors beyond the early developmental stages and provide mechanistic links between mitochondrial dysfunctions, α-synuclein aggregation, and the survival of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons/metabolism , LIM-Homeodomain Proteins/genetics , Mesencephalon/metabolism , Mitochondria/metabolism , Transcription Factors/genetics , Animals , Cell Survival/genetics , DNA Damage , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , LIM-Homeodomain Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Oxidative Stress , Protein Aggregation, Pathological , Transcription Factors/deficiency , alpha-Synuclein/metabolism
14.
Bioorg Med Chem Lett ; 25(15): 2958-62, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26048804

ABSTRACT

Molecular modeling was performed on a triazolo quinazoline lead compound to help develop a series of adenosine A2A receptor antagonists with improved hERG profile. Superposition of the lead compound onto MK-499, a benchmark hERG inhibitor, combined with pKa calculations and measurement, identified terminal fluorobenzene to be responsible for hERG activity. Docking of the lead compound into an A2A crystal structure suggested that this group is located at a flexible, spacious, and solvent-exposed opening of the binding pocket, making it possible to tolerate various functional groups. Transformation analysis (MMP, matched molecular pair) of in-house available experimental data on hERG provided suggestions for modifications in order to mitigate this liability. This led to the synthesis of a series of compounds with significantly reduced hERG activity. The strategy used in the modeling work can be applied to other medicinal chemistry programs to help improve hERG profile.


Subject(s)
Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Ether-A-Go-Go Potassium Channels/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Receptor, Adenosine A2A/metabolism , Benzopyrans/chemistry , Benzopyrans/pharmacology , Drug Design , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Humans , Molecular Docking Simulation , Piperidines/chemistry , Piperidines/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
15.
Development ; 138(16): 3399-408, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21752929

ABSTRACT

The severe disorders associated with a loss or dysfunction of midbrain dopamine neurons (DNs) have intensified research aimed at deciphering developmental programs controlling midbrain development. The homeodomain proteins Lmx1a and Lmx1b are important for the specification of DNs during embryogenesis, but it is unclear to what degree they may mediate redundant or specific functions. Here, we provide evidence showing that DN progenitors in the ventral midbrain can be subdivided into molecularly distinct medial and lateral domains, and these subgroups show different sensitivity to the loss of Lmx1a and Lmx1b. Lmx1a is specifically required for converting non-neuronal floor-plate cells into neuronal DN progenitors, a process that involves the establishment of Notch signaling in ventral midline cells. On the other hand, lateral DN progenitors that do not appear to originate from the floor plate are selectively ablated in Lmx1b mutants. In addition, we also reveal an unanticipated role for Lmx1b in regulating Phox2a expression and the sequential specification of ocular motor neurons (OMNs) and red nucleus neurons (RNNs) from progenitors located lateral to DNs in the midbrain. Our data therefore establish that Lmx1b influences the differentiation of multiple neuronal subtypes in the ventral midbrain, whereas Lmx1a appears to be exclusively devoted to the differentiation of the DN lineage.


Subject(s)
Homeodomain Proteins/metabolism , Mesencephalon/embryology , Mesencephalon/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Cell Lineage , Dopamine/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Receptors, Notch/metabolism , Signal Transduction , Transcription Factors/genetics
16.
Hum Reprod Update ; 30(4): 410-441, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38478759

ABSTRACT

BACKGROUND: With increasing significance of developmental programming effects associated with placental dysfunction, more investigations are devoted to improving the characterization and understanding of placental signatures in health and disease. The placenta is a transitory but dynamic organ adapting to the shifting demands of fetal development and available resources of the maternal supply throughout pregnancy. Trophoblasts (cytotrophoblasts, syncytiotrophoblasts, and extravillous trophoblasts) are placental-specific cell types responsible for the main placental exchanges and adaptations. Transcriptomic studies with single-cell resolution have led to advances in understanding the placenta's role in health and disease. These studies, however, often show discrepancies in characterization of the different placental cell types. OBJECTIVE AND RATIONALE: We aim to review the knowledge regarding placental structure and function gained from the use of single-cell RNA sequencing (scRNAseq), followed by comparing cell-type-specific genes, highlighting their similarities and differences. Moreover, we intend to identify consensus marker genes for the various trophoblast cell types across studies. Finally, we will discuss the contributions and potential applications of scRNAseq in studying pregnancy-related diseases. SEARCH METHODS: We conducted a comprehensive systematic literature review to identify different cell types and their functions at the human maternal-fetal interface, focusing on all original scRNAseq studies on placentas published before March 2023 and published reviews (total of 28 studies identified) using PubMed search. Our approach involved curating cell types and subtypes that had previously been defined using scRNAseq and comparing the genes used as markers or identified as potential new markers. Next, we reanalyzed expression matrices from the six available scRNAseq raw datasets with cell annotations (four from first trimester and two at term), using Wilcoxon rank-sum tests to compare gene expression among studies and annotate trophoblast cell markers in both first trimester and term placentas. Furthermore, we integrated scRNAseq raw data available from 18 healthy first trimester and nine term placentas, and performed clustering and differential gene expression analysis. We further compared markers obtained with the analysis of annotated and raw datasets with the literature to obtain a common signature gene list for major placental cell types. OUTCOMES: Variations in the sampling site, gestational age, fetal sex, and subsequent sequencing and analysis methods were observed between the studies. Although their proportions varied, the three trophoblast types were consistently identified across all scRNAseq studies, unlike other non-trophoblast cell types. Notably, no marker genes were shared by all studies for any of the investigated cell types. Moreover, most of the newly defined markers in one study were not observed in other studies. These discrepancies were confirmed by our analysis on trophoblast cell types, where hundreds of potential marker genes were identified in each study but with little overlap across studies. From 35 461 and 23 378 cells of high quality in the first trimester and term placentas, respectively, we obtained major placental cell types, including perivascular cells that previously had not been identified in the first trimester. Importantly, our meta-analysis provides marker genes for major placental cell types based on our extensive curation. WIDER IMPLICATIONS: This review and meta-analysis emphasizes the need for establishing a consensus for annotating placental cell types from scRNAseq data. The marker genes identified here can be deployed for defining human placental cell types, thereby facilitating and improving the reproducibility of trophoblast cell annotation.


Subject(s)
Placenta , Sequence Analysis, RNA , Single-Cell Analysis , Trophoblasts , Humans , Female , Pregnancy , Placenta/metabolism , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Trophoblasts/metabolism , Transcriptome
17.
Dev Cell ; 59(10): 1269-1283.e6, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38565145

ABSTRACT

Progenitor cells adapt their behavior in response to tissue demands. However, the molecular mechanisms controlling esophageal progenitor decisions remain largely unknown. Here, we demonstrate the presence of a Troy (Tnfrsf19)-expressing progenitor subpopulation localized to defined regions along the mouse esophageal axis. Lineage tracing and mathematical modeling demonstrate that Troy-positive progenitor cells are prone to undergoing symmetrical fate choices and contribute to esophageal tissue homeostasis long term. Functionally, TROY inhibits progenitor proliferation and enables commitment to differentiation without affecting fate symmetry. Whereas Troy expression is stable during esophageal homeostasis, progenitor cells downregulate Troy in response to tissue stress, enabling proliferative expansion of basal cells refractory to differentiation and reestablishment of tissue homeostasis. Our results demonstrate functional, spatially restricted progenitor heterogeneity in the esophageal epithelium and identify how dynamic regulation of Troy coordinates tissue generation.


Subject(s)
Cell Differentiation , Cell Proliferation , Esophagus , Receptors, Tumor Necrosis Factor , Stem Cells , Animals , Mice , Cell Lineage , Epithelium/metabolism , Esophageal Mucosa/metabolism , Esophageal Mucosa/cytology , Esophagus/cytology , Esophagus/metabolism , Homeodomain Proteins , Homeostasis , Stem Cells/metabolism , Stem Cells/cytology , Receptors, Tumor Necrosis Factor/analysis , Receptors, Tumor Necrosis Factor/metabolism
18.
Cell Rep Med ; 5(5): 101515, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38631348

ABSTRACT

During pregnancy, germline development is vital for maintaining the continuation of species. Recent studies have shown increased pregnancy risks in COVID-19 patients at the perinatal stage. However, the potential consequence of infection for reproductive quality in developing fetuses remains unclear. Here, we analyze the transcriptome and DNA methylome of the fetal germline following maternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We find that infection at early gestational age, a critical period of human primordial germ cell specification and epigenetic reprogramming, trivially affects fetal germ cell (FGC) development. Additionally, FGC-niche communications are not compromised by maternal infection. Strikingly, both general and SARS-CoV-2-specific immune pathways are greatly activated in gonadal niche cells to protect FGCs from maternal infection. Notably, there occurs an "in advance" development tendency in FGCs after maternal infection. Our study provides insights into the impacts of maternal SARS-CoV-2 infection on fetal germline development and serves as potential clinical guidance for future pandemics.


Subject(s)
COVID-19 , Fetus , Germ Cells , SARS-CoV-2 , Humans , Female , COVID-19/virology , COVID-19/immunology , COVID-19/pathology , Pregnancy , Germ Cells/virology , Fetus/virology , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/pathology , Gonads/virology , Transcriptome/genetics , Male , DNA Methylation/genetics , Epigenesis, Genetic
19.
Commun Biol ; 6(1): 69, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36653487

ABSTRACT

Excessive androgen production and obesity are key to polycystic ovary syndrome (PCOS) pathogenesis. Prenatal androgenized (PNA), peripubertal androgenized, and overexpression of nerve growth factor in theca cells (17NF) are commonly used PCOS-like mouse models and diet-induced maternal obesity model is often included for comparsion. To reveal the molecular features of these models, we have performed transcriptome survey of the hypothalamus, adipose tissue, ovary and metaphase II (MII) oocytes. The largest number of differentially expressed genes (DEGs) is found in the ovaries of 17NF and in the adipose tissues of peripubertal androgenized models. In contrast, hypothalamus is most affected in PNA and maternal obesity models suggesting fetal programming effects. The Ms4a6e gene, membrane-spanning 4-domains subfamily A member 6E, a DEG identified in the adipose tissue in all mouse models is also differently expressed in adipose tissue of women with PCOS, highlighting a conserved disease function. Our comprehensive transcriptomic profiling of key target tissues involved in PCOS pathology highlights the effects of developmental windows for androgen exposure and maternal obesity, and provides unique resource to investigate molecular mechanisms underlying PCOS pathogenesis.


Subject(s)
Obesity, Maternal , Polycystic Ovary Syndrome , Mice , Animals , Female , Pregnancy , Humans , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Androgens/metabolism , Transcriptome , Obesity, Maternal/complications
20.
Cell Rep ; 42(1): 112021, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36848234

ABSTRACT

Different formative pluripotent stem cells harboring similar functional properties have been recently established to be lineage neutral and germline competent yet have distinct molecular identities. Here, we show that WNT/ß-catenin signaling activation sustains transient mouse epiblast-like cells as epiblast-like stem cells (EpiLSCs). EpiLSCs display metastable formative pluripotency with bivalent cellular energy metabolism and unique transcriptomic features and chromatin accessibility. We develop single-cell stage label transfer (scSTALT) to study the formative pluripotency continuum and reveal that EpiLSCs recapitulate a unique developmental period in vivo, filling the gap of the formative pluripotency continuum between other published formative stem cells. WNT/ß-catenin signaling activation counteracts differentiation effects of activin A and bFGF by preventing complete dissolution of naive pluripotency regulatory network. Moreover, EpiLSCs have direct competence toward germline specification, which is further matured by an FGF receptor inhibitor. Our EpiLSCs can serve as an in vitro model for mimicking and studying early post-implantation development and pluripotency transition.


Subject(s)
Pluripotent Stem Cells , Wnt Signaling Pathway , Animals , Mice , beta Catenin , Cell Differentiation , Germ Cells , Germ Layers
SELECTION OF CITATIONS
SEARCH DETAIL