Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 513
Filter
1.
Article in English | MEDLINE | ID: mdl-35134541

ABSTRACT

To explore the developmental toxicity of cefixime (CE) in the developmental disorder and toxicity mechanism of CE on otic vesicles, zebrafish embryos were used as an animal model. The results showed that CE increased mortality in a dose-dependent manner and decreased the hatching rate of zebrafish larva at 96 hpf. Interestingly, CE significantly reduced the area of the saccule and utricle, as well as the area of otic vesicles in zebrafish larvae (p < 0.001). Fibroblast growth factor 8a (Fgf8a) inhibitors and bone morphogenetic protein (BMP) inhibitors caused similar morphological changes. CE decreased the lateral hair cells of zebrafish larvae in a dose-dependent manner. Furthermore, CE caused the downregulation of cartilage and bone-related genes and Na+/K+-ATPase-related genes of zebrafish larvae at 72 hpf and 120 hpf according to RT-qPCR. A comparison with the control group revealed that 100 µg/mL CE also caused a decrease in Na+/K+-ATPase activity (p < 0.01). In addition, antibody staining verified that CE inhibited the expression of Na+/K+-ATPase in the otic vesicles and the nephridium of zebrafish larvae. The data obtained in this study suggested that CE has significant ototoxicity during embryonic development of zebrafish, which is closely related to Na+/K+-ATPase and the regulation of the Fgf8a/BMP signaling pathways. The effects and toxicity of CE on ear development in other animal models need to be further explored.


Subject(s)
Cefixime/toxicity , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Gene Expression Regulation, Developmental/drug effects , Organogenesis/drug effects , Animals , Anti-Bacterial Agents/toxicity , Larva/drug effects , Zebrafish
2.
Nat Protoc ; 16(12): 5652-5672, 2021 12.
Article in English | MEDLINE | ID: mdl-34759383

ABSTRACT

Heart-forming organoids (HFOs) derived from human pluripotent stem cells (hPSCs) are a complex, highly structured in vitro model of early heart, foregut and vasculature development. The model represents a potent tool for various applications, including teratogenicity studies, gene function analysis and drug discovery. Here, we provide a detailed protocol describing how to form HFOs within 14 d. In an initial 4 d preculture period, hPSC aggregates are individually formed in a 96-well format and then Matrigel-embedded. Subsequently, the chemical WNT pathway modulators CHIR99021 and IWP2 are applied, inducing directed differentiation. This highly robust protocol can be used on many different hPSC lines and be combined with manipulation technologies such as gene targeting and drug testing. HFO formation can be assessed by numerous complementary methods, ranging from various imaging approaches to gene expression studies. Here, we highlight the flow cytometry-based analysis of individual HFOs, enabling the quantitative monitoring of lineage formation.


Subject(s)
Flow Cytometry/methods , Organogenesis/genetics , Organoids/cytology , Pluripotent Stem Cells/cytology , Tissue Scaffolds , Wnt Signaling Pathway/drug effects , Benzothiazoles/pharmacology , Cell Differentiation/drug effects , Collagen/chemistry , Collagen/pharmacology , Drug Combinations , Drug Discovery/methods , Gene Targeting/methods , Heart/diagnostic imaging , Heart/drug effects , Humans , Laminin/chemistry , Laminin/pharmacology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Organogenesis/drug effects , Organoids/diagnostic imaging , Organoids/drug effects , Organoids/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proteoglycans/chemistry , Proteoglycans/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Teratogens/toxicity
3.
Dev Cell ; 56(22): 3128-3145.e15, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34762852

ABSTRACT

Identification of physiological modulators of nuclear hormone receptor (NHR) activity is paramount for understanding the link between metabolism and transcriptional networks that orchestrate development and cellular physiology. Using libraries of metabolic enzymes alongside their substrates and products, we identify 1-deoxysphingosines as modulators of the activity of NR2F1 and 2 (COUP-TFs), which are orphan NHRs that are critical for development of the nervous system, heart, veins, and lymphatic vessels. We show that these non-canonical alanine-based sphingolipids bind to the NR2F1/2 ligand-binding domains (LBDs) and modulate their transcriptional activity in cell-based assays at physiological concentrations. Furthermore, inhibition of sphingolipid biosynthesis phenocopies NR2F1/2 deficiency in endothelium and cardiomyocytes, and increases in 1-deoxysphingosine levels activate NR2F1/2-dependent differentiation programs. Our findings suggest that 1-deoxysphingosines are physiological regulators of NR2F1/2-mediated transcription.


Subject(s)
Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Organogenesis/drug effects , Sphingolipids/pharmacology , Animals , Cell Differentiation/physiology , Gene Expression Regulation/physiology , Humans , Lymphatic Vessels/drug effects , Mice , Organogenesis/physiology , Repressor Proteins/physiology
4.
Cells ; 10(10)2021 10 09.
Article in English | MEDLINE | ID: mdl-34685691

ABSTRACT

Seizure threshold 2 (SZT2) is a component of the KICSTOR complex which, under catabolic conditions, functions as a negative regulator in the amino acid-sensing branch of mTORC1. Mutations in this gene cause a severe neurodevelopmental and epileptic encephalopathy whose main symptoms include epilepsy, intellectual disability, and macrocephaly. As SZT2 remains one of the least characterized regulators of mTORC1, in this work we performed a systematic interactome analysis under catabolic and anabolic conditions. Besides numerous mTORC1 and AMPK signaling components, we identified clusters of proteins related to autophagy, ciliogenesis regulation, neurogenesis, and neurodegenerative processes. Moreover, analysis of SZT2 ablated cells revealed increased mTORC1 signaling activation that could be reversed by Rapamycin or Torin treatments. Strikingly, SZT2 KO cells also exhibited higher levels of autophagic components, independent of the physiological conditions tested. These results are consistent with our interactome data, in which we detected an enriched pool of selective autophagy receptors/regulators. Moreover, preliminary analyses indicated that SZT2 alters ciliogenesis. Overall, the data presented form the basis to comprehensively investigate the physiological functions of SZT2 that could explain major molecular events in the pathophysiology of developmental and epileptic encephalopathy in patients with SZT2 mutations.


Subject(s)
Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Protein Interaction Maps , Amino Acids/deficiency , Animals , Blood Proteins/pharmacology , Cilia/drug effects , Cilia/metabolism , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Organogenesis/drug effects , Principal Component Analysis , Protein Interaction Maps/drug effects , Sirolimus/pharmacology
5.
J Cell Mol Med ; 25(19): 9447-9459, 2021 10.
Article in English | MEDLINE | ID: mdl-34498380

ABSTRACT

Oestradiol (E2) is a critical factor for multiple systems' development during the embryonic period. Here, we aimed to investigate the effects of oestradiol on intrahepatic bile duct development, which may allow a better understanding of congenital bile duct dysplasia. DLK+ hepatoblasts were extracted from the C57BL/6CrSlc foetal mice and randomly divided into control group, oestradiol groups (1, 10, 100 nM) and oestradiol (10 nM) + DAPT (inhibitor of Notch signalling; 40 µM) group for in vitro experiments. For in vivo analysis, pregnant mice were divided into control group, oestradiol (intraperitoneal injection of 0.6 mg/kg/day) ± DAPT (subcutaneous injection of 10 mg/kg/day) groups and tamoxifen (gavage administration of 0.4 mg/kg/day) group. The results showed that oestradiol promoted hepatoblast differentiation into cholangiocytes and intrahepatic bile duct development during the embryonic period. Tamoxifen, an antioestrogenic drug, inhibited the above processes. Moreover, oestradiol promoted the expression of Notch signalling pathway-associated proteins and genes both in vitro and in vivo. Notably, DAPT addition inhibited the oestradiol-mediated effects. In conclusion, oestradiol can promote hepatoblast differentiation into cholangiocytes and intrahepatic bile duct development of C57BL/6CrSlc mice during embryonic period via the Notch signalling pathway.


Subject(s)
Bile Ducts, Intrahepatic/embryology , Bile Ducts, Intrahepatic/metabolism , Estradiol/metabolism , Organogenesis , Receptors, Notch/metabolism , Signal Transduction , Animals , Biomarkers , Cell Differentiation , Cells, Cultured , Estradiol/pharmacology , Gene Expression , Hepatocytes/metabolism , Immunohistochemistry , Immunophenotyping , Mice , Mice, Inbred C57BL , Organogenesis/drug effects , Stem Cells/metabolism
6.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502408

ABSTRACT

Oxidative stress plays a pathological role in pulmonary hypoplasia and pulmonary hypertension in congenital diaphragmatic hernia (CDH). This study investigated the effect of molecular hydrogen (H2), an antioxidant, on CDH pathology induced by nitrofen. Sprague-Dawley rats were divided into three groups: control, CDH, and CDH + hydrogen-rich water (HW). Pregnant dams of CDH + HW pups were orally administered HW from embryonic day 10 until parturition. Gasometric evaluation and histological, immunohistochemical, and real-time polymerase chain reaction analyses were performed. Gasometric results (pH, pO2, and pCO2 levels) were better in the CDH + HW group than in the CDH group. The CDH + HW group showed amelioration of alveolarization and pulmonary artery remodeling compared with the CDH group. Oxidative stress (8-hydroxy-2'-deoxyguanosine-positive-cell score) in the pulmonary arteries and mRNA levels of protein-containing pulmonary surfactant that protects against pulmonary collapse (surfactant protein A) were significantly attenuated in the CDH + HW group compared with the CDH group. Overall, prenatal H2 administration improved respiratory function by attenuating lung morphology and pulmonary artery thickening in CDH rat models. Thus, H2 administration in pregnant women with diagnosed fetal CDH might be a novel antenatal intervention strategy to reduce newborn mortality due to CDH.


Subject(s)
Hernias, Diaphragmatic, Congenital/drug therapy , Hydrogen/pharmacology , Animals , Animals, Newborn , Antioxidants/pharmacology , Deuterium Oxide/pharmacology , Disease Models, Animal , Female , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/pathology , Hydrogen/metabolism , Hypertension, Pulmonary/metabolism , Lung/pathology , Male , Organogenesis/drug effects , Phenyl Ethers/adverse effects , Phenyl Ethers/pharmacology , Pregnancy , Pulmonary Artery , Pulmonary Surfactants/metabolism , Rats , Rats, Sprague-Dawley , Vascular Remodeling/drug effects
7.
Int J Mol Sci ; 22(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34576143

ABSTRACT

Nitro-oleic acid (NO2-OA), pluripotent cell-signaling mediator, was recently described as a modulator of the signal transducer and activator of transcription 3 (STAT3) activity. In our study, we discovered new aspects of NO2-OA involvement in the regulation of stem cell pluripotency and differentiation. Murine embryonic stem cells (mESC) or mESC-derived embryoid bodies (EBs) were exposed to NO2-OA or oleic acid (OA) for selected time periods. Our results showed that NO2-OA but not OA caused the loss of pluripotency of mESC cultivated in leukemia inhibitory factor (LIF) rich medium via the decrease of pluripotency markers (NANOG, sex-determining region Y-box 1 transcription factor (SOX2), and octamer-binding transcription factor 4 (OCT4)). The effects of NO2-OA on mESC correlated with reduced phosphorylation of STAT3. Subsequent differentiation led to an increase of the ectodermal marker orthodenticle homolog 2 (Otx2). Similarly, treatment of mESC-derived EBs by NO2-OA resulted in the up-regulation of both neural markers Nestin and ß-Tubulin class III (Tubb3). Interestingly, the expression of cardiac-specific genes and beating of EBs were significantly decreased. In conclusion, NO2-OA is able to modulate pluripotency of mESC via the regulation of STAT3 phosphorylation. Further, it attenuates cardiac differentiation on the one hand, and on the other hand, it directs mESC into neural fate.


Subject(s)
Cell Differentiation , Mouse Embryonic Stem Cells/cytology , Neurons/cytology , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Mice , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neurons/drug effects , Neurons/metabolism , Organogenesis/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
8.
Development ; 148(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34142711

ABSTRACT

Axial elongation of the neural tube is crucial during mammalian embryogenesis for anterior-posterior body axis establishment and subsequent spinal cord development, but these processes cannot be interrogated directly in humans as they occur post-implantation. Here, we report an organoid model of neural tube extension derived from human pluripotent stem cell (hPSC) aggregates that have been caudalized with Wnt agonism, enabling them to recapitulate aspects of the morphological and temporal gene expression patterns of neural tube development. Elongating organoids consist largely of neuroepithelial compartments and contain TBXT+SOX2+ neuro-mesodermal progenitors in addition to PAX6+NES+ neural progenitors. A critical threshold of Wnt agonism stimulated singular axial extensions while maintaining multiple cell lineages, such that organoids displayed regionalized anterior-to-posterior HOX gene expression with hindbrain (HOXB1) regions spatially distinct from brachial (HOXC6) and thoracic (HOXB9) regions. CRISPR interference-mediated silencing of TBXT, a Wnt pathway target, increased neuroepithelial compartmentalization, abrogated HOX expression and disrupted uniaxial elongation. Together, these results demonstrate the potent capacity of caudalized hPSC organoids to undergo axial elongation in a manner that can be used to dissect the cellular organization and patterning decisions that dictate early human nervous system development.


Subject(s)
Body Patterning , Neural Tube/embryology , Organogenesis , Organoids , Body Patterning/drug effects , Cell Differentiation , Embryonic Development , Gene Expression Regulation, Developmental , Humans , Mesoderm/embryology , Mesoderm/metabolism , Neurogenesis/drug effects , Organogenesis/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway/drug effects
9.
FASEB J ; 35(7): e21718, 2021 07.
Article in English | MEDLINE | ID: mdl-34105801

ABSTRACT

Acetaminophen, aspirin, and ibuprofen are mild analgesics commonly used by pregnant women, the sole current recommendation being to avoid ibuprofen from the fifth month of gestation. The nephrotoxicity of these three analgesics is well documented in adults, as is their interference with prostaglandins biosynthesis. Here we investigated the effect of these analgesics on human first trimester kidneys ex vivo. We first evaluated prostaglandins biosynthesis functionality by performing a wide screening of prostaglandin expression patterns in first trimester human kidneys. We demonstrated that prostaglandins biosynthesis machinery is functional during early nephrogenesis. Human fetal kidney explants aged 7-12 developmental weeks were exposed ex vivo to ibuprofen, aspirin or acetaminophen for 7 days, and analyzed by histology, immunohistochemistry, and flow cytometry. This study has revealed that these analgesics induced a spectrum of abnormalities within early developing structures, ranging from cell death to a decline in differentiating glomeruli density. These results warrant caution for the use of these medicines during the first trimester of pregnancy.


Subject(s)
Analgesics/adverse effects , Fetus/drug effects , Kidney Glomerulus/drug effects , Organogenesis/drug effects , Cell Death/drug effects , Female , Fetus/metabolism , Humans , Kidney Glomerulus/metabolism , Pregnancy , Pregnancy Trimester, First/drug effects , Prostaglandins/metabolism
10.
Dev Dyn ; 250(12): 1759-1777, 2021 12.
Article in English | MEDLINE | ID: mdl-34056790

ABSTRACT

BACKGROUND: Biomechanical stimuli are known to be important to cardiac development, but the mechanisms are not fully understood. Here, we pharmacologically disrupted the biomechanical environment of wild-type zebrafish embryonic hearts for an extended duration and investigated the consequent effects on cardiac function, morphological development, and gene expression. RESULTS: Myocardial contractility was significantly diminished or abolished in zebrafish embryonic hearts treated for 72 hours from 2 dpf with 2,3-butanedione monoxime (BDM). Image-based flow simulations showed that flow wall shear stresses were abolished or significantly reduced with high oscillatory shear indices. At 5 dpf, after removal of BDM, treated embryonic hearts were maldeveloped, having disrupted cardiac looping, smaller ventricles, and poor cardiac function (lower ejected flow, bulboventricular regurgitation, lower contractility, and slower heart rate). RNA sequencing of cardiomyocytes of treated hearts revealed 922 significantly up-regulated genes and 1,698 significantly down-regulated genes. RNA analysis and subsequent qPCR and histology validation suggested that biomechanical disruption led to an up-regulation of inflammatory and apoptotic genes and down-regulation of ECM remodeling and ECM-receptor interaction genes. Biomechanics disruption also prevented the formation of ventricular trabeculation along with notch1 and erbb4a down-regulation. CONCLUSIONS: Extended disruption of biomechanical stimuli caused maldevelopment, and potential genes responsible for this are identified.


Subject(s)
Biomechanical Phenomena/drug effects , Diacetyl/analogs & derivatives , Heart/embryology , Zebrafish , Animals , Animals, Genetically Modified , Biomechanical Phenomena/physiology , Diacetyl/pharmacology , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Embryonic Development/genetics , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Heart/physiology , Hydrodynamics , Myocardial Contraction/drug effects , Myocardium/metabolism , Organogenesis/drug effects , Organogenesis/genetics , Organogenesis/physiology , Stress, Mechanical , Zebrafish/embryology , Zebrafish/genetics
11.
Toxicol Ind Health ; 37(7): 391-397, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34047658

ABSTRACT

Di-2-ethylhexyl phthalate (DEHP) is a type of plasticizer widely used in industry. It is well-known for its toxic effects to endocrine and reproductive systems and has been detected in amniotic fluid and placenta. In the present study, we explored the effects of DEHP on heart development by using zebrafish as a model organism. DEHP (0.02 pg) was injected into the yolk sac of zebrafish embryos at the one-cell stage. No significant difference was found in embryonic lethality between control and DEHP groups at 1-day postfertilization (dpf), but mortality significantly increased in DEHP groups at 2 and 3 dpf. The average heart rate was significantly reduced in the surviving DEHP-treated zebrafish larvae at 3 and 4 dpf. In addition, massive pericardial edema was found in DEHP-treated zebrafish (12.6 ± 1.5%), which was significantly higher than that of the control group. Serious heart looping disorder was also observed in DEHP-treated larvae, mainly manifested with an elongated atrial-ventricular distance. Moreover, the expression of heart development transcription factors was affected by DEHP injection. Real-time polymerase chain reaction confirmed that five transcription factors (hand2, tp53, mef2c, esr1, and tbx18) were significantly downregulated in the DEHP group at 2 dpf, and three transcription factors (zic3, tcf21, and gata4) were significantly upregulated. Our results emphasize the need for the development of a nontoxic plasticizer to prevent possible deleterious effects on humans and other life-forms.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/drug effects , Diethylhexyl Phthalate/toxicity , Embryo, Nonmammalian/drug effects , Heart/drug effects , Heart/growth & development , Membrane Glycoproteins/drug effects , Zebrafish Proteins/drug effects , Zebrafish/growth & development , Animals , Embryonic Development/drug effects , Humans , Occupational Exposure/adverse effects , Organogenesis/drug effects , Plasticizers/toxicity , Water Pollutants, Chemical/toxicity
12.
Stem Cell Reports ; 16(5): 1143-1155, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33891872

ABSTRACT

When stimulated with a pulse from an exogenous WNT pathway activator, small aggregates of mouse embryonic stem cells (ESCs) can undergo embryo-like axial morphogenesis and patterning along the three major body axes. However, these structures, called gastruloids, currently lack the anterior embryonic regions, such as those belonging to the brain. Here, we describe an approach to generate gastruloids that have a more complete antero-posterior development. We used hydrogel microwell arrays to promote the robust derivation of mouse ESCs into post-implantation epiblast-like (EPI) aggregates in a reproducible and scalable manner. These EPI aggregates break symmetry and axially elongate without external chemical stimulation. Inhibition of WNT signaling in early stages of development leads to the formation of gastruloids with anterior neural tissues. Thus, we provide a new tool to study the development of the mouse after implantation in vitro, especially the formation of anterior neural regions.


Subject(s)
Body Patterning , Gastrula/growth & development , Nerve Tissue/growth & development , Organogenesis , Wnt Proteins/metabolism , Animals , Body Patterning/drug effects , Cell Aggregation/drug effects , Cell Line , Gastrula/drug effects , Germ Layers/cytology , Germ Layers/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Hydrogels/pharmacology , Mice , Nerve Tissue/drug effects , Organogenesis/drug effects , Polyethylene Glycols/pharmacology , Wnt Signaling Pathway/drug effects
13.
Reprod Toxicol ; 102: 43-55, 2021 06.
Article in English | MEDLINE | ID: mdl-33848595

ABSTRACT

The brominated flame retardants (BFRs), 1,2-dibromo-4-(1,2 dibromoethyl)cyclohexane (TBECH) and 2,3-dibromopropyl-2,4,6-tribromophenyl ether (DPTE) bind to the androgen receptor (AR). in vitro bioassays have shown that TBECH is a potent androgen agonist while DPTE is a potent AR antagonist. Both TBECH and DPTE alter gene expression associated with AR regulation. However, it remains to be determined if TBECH and DPTE can affect the prostate. For this reason, we exposed CD1 mice to a 1:1 mixture of TBECH diastereomers α and ß, a 1:1 mixture of γ and δ, and to DPTE, and tested their effects on prostate growth, histology and gene expression profiles. Castrated mice were used to study the androgenic effects of TBECHαß and TBECHγδ while the antagonistic effects of DPTE were studied in non-castrated mice. We observed that testosterone and TBECHγδ increased body and prostate weights while TBECHαß affected neither of them; and that DPTE had no effect on body weight but reduced prostate weight drastically. Histomorphometric analysis of the prostate revealed epithelial and glandular alterations in the TBECHγδ group comparable to those in testosterone group while alterations in the TBECHαß group were less pronounced. DPTE displayed androgen antagonist activity reminiscent of castration. The transcription profile of the prostate was altered by castration and exposure to testosterone and to TBECHγδ reversed several of these changes. Testosterone and TBECHγδ also regulated the expression of several androgen responsive genes implicated in prostate growth and cancer. While DPTE resulted in a drastic reduction in prostate weight, it only affected a small number of genes. The results indicate that TBECHγδ and DPTE are of high human health concern as they may contribute to changes in prostate growth, histology and function.


Subject(s)
Cyclohexanes/toxicity , Endocrine Disruptors/toxicity , Flame Retardants/toxicity , Hydrocarbons, Brominated/toxicity , Prostate/drug effects , Androgen Antagonists , Androgen Receptor Antagonists , Androgens , Animals , Cell Line, Tumor , Endocrine Disruptors/metabolism , Gene Expression/drug effects , Halogenation , Humans , Male , Mice , Organogenesis/drug effects , Prostate/growth & development , Prostate/metabolism , Receptors, Androgen/metabolism
14.
Stem Cell Reports ; 16(5): 1262-1275, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33836146

ABSTRACT

Despite heterogeneity across the six layers of the mammalian cortex, all excitatory neurons are generated from a single founder population of neuroepithelial stem cells. However, how these progenitors alter their layer competence over time remains unknown. Here, we used human embryonic stem cell-derived cortical progenitors to examine the role of fibroblast growth factor (FGF) and Notch signaling in influencing cell fate, assessing their impact on progenitor phenotype, cell-cycle kinetics, and layer specificity. Forced early cell-cycle exit, via Notch inhibition, caused rapid, near-exclusive generation of deep-layer VI neurons. In contrast, prolonged FGF2 promoted proliferation and maintained progenitor identity, delaying laminar progression via MAPK-dependent mechanisms. Inhibiting MAPK extended cell-cycle length and led to generation of layer-V CTIP2+ neurons by repressing alternative laminar fates. Taken together, FGF/MAPK regulates the proliferative/neurogenic balance in deep-layer corticogenesis and provides a resource for generating layer-specific neurons for studying development and disease.


Subject(s)
Cerebral Cortex/embryology , Fibroblast Growth Factors/metabolism , Mitogen-Activated Protein Kinases/metabolism , Organogenesis , Signal Transduction , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Gene Regulatory Networks/drug effects , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organogenesis/drug effects , PAX6 Transcription Factor/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Tumor Suppressor Proteins/metabolism
15.
Front Endocrinol (Lausanne) ; 12: 609308, 2021.
Article in English | MEDLINE | ID: mdl-33716967

ABSTRACT

Thyroid hormones mediate a remarkable range of functions in many tissues and organ systems through the thyroid hormone receptors-THRA and THRB. Tissues and organs are composed of heterogeneous cells of different cell types. These different cell types have varying receptor expression abilities, which lead to variable responses in thyroid hormone regulation. The tissue-specific Thra and Thrb gene expression patterns help us understand the action of thyroid hormones at the tissue level. However, the situation becomes complicated if we wish to focus on tissues more closely to trace the responsive cells, which is a vital step in the process of understanding the molecular mechanism of diseases related to thyroid hormone regulation. Single-cell RNA sequencing technology is a powerful tool used to profile gene expression programs in individual cells. The Tabula Muris Consortium generates a single-cell transcriptomic atlas across the life span of Mus musculus that includes data from 23 tissues and organs. It provides an unprecedented opportunity to understand thyroid hormone regulation at the cell type resolution. We demonstrated the approaches that allow application of the single-cell RNA-Seq data generated by the Tabula Muris Consortium to trace responsive cells in tissues. First, employing the single-cell RNA-Seq data, we calculated the ability of different cell types to express Thra and Thrb, which direct us to the cell types sensitive to thyroid hormone regulation in tissues and organs. Next, using a cell clustering algorithm, we explored the subtypes with low Thra or Thrb expression within the different cell types and identified the potentially responsive cell subtypes. Finally, in the liver tissue treated with thyroid hormones, using the single-cell RNA-Seq data, we successfully traced the responsive cell types. We acknowledge that the computational predictions reported here need to be further validated using wet-lab experiments. However, we believe our results provide powerful information and will be beneficial for wet lab researchers.


Subject(s)
Organ Specificity , RNA-Seq/statistics & numerical data , Single-Cell Analysis/statistics & numerical data , Triiodothyronine/pharmacology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Lineage/drug effects , Cell Lineage/genetics , Computational Biology , Data Analysis , Female , Gene Expression/drug effects , Male , Mice , Mice, Inbred C57BL , Organ Specificity/drug effects , Organ Specificity/genetics , Organogenesis/drug effects , Organogenesis/genetics , RNA-Seq/methods , Receptors, Thyroid Hormone/genetics , Single-Cell Analysis/methods , Thyroid Hormones/pharmacology
16.
Differentiation ; 118: 34-40, 2021.
Article in English | MEDLINE | ID: mdl-33707128

ABSTRACT

The development of the female reproductive tract can be divided into three parts consisting of Müllerian duct organogenesis, pre-sexual maturation organ development, and post-sexual maturation hormonal regulation. In primates, Müllerian duct organogenesis proceeds in an estrogen independent fashion based on transcriptional pathways that are suppressed in males by the presence of AMH and SRY. However, clinical experience indicates that exposure to xenoestrogens such as diethylstilbestrol (DES) during critical periods including late organogenesis and pre-sexual maturational development can have substantial effects on uterine morphology, and confer increased risk of disease states later in life. Recent evidence has demonstrated that these effects are in part due to epigenetic regulation of gene expression, both in the form of aberrant CpG methylation, and accompanying histone modifications. While xenoestrogens and selective estrogen receptor modulators (SERMS) both can induce non-canonical binding confirmations in estrogen receptors, the primate specific fetal estrogens Estriol and Estetrol may act in a similar fashion to alter gene expression through tissue specific epigenetic modulation.


Subject(s)
DNA Methylation/genetics , Estrogens/genetics , Genitalia, Female/growth & development , Organogenesis/genetics , Animals , DNA Methylation/drug effects , Diethylstilbestrol/pharmacology , Epigenesis, Genetic/genetics , Estradiol/metabolism , Estrogens/metabolism , Female , Genitalia, Female/metabolism , Humans , Mullerian Ducts/drug effects , Mullerian Ducts/growth & development , Mullerian Ducts/metabolism , Organogenesis/drug effects , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Uterus/drug effects , Uterus/growth & development
17.
Reprod Toxicol ; 101: 81-92, 2021 04.
Article in English | MEDLINE | ID: mdl-33713778

ABSTRACT

Redox regulation during metazoan development ensures that coordinated metabolic reprogramming and developmental signaling are orchestrated with high fidelity in the hypoxic embryonic environment. Valproic acid (VPA), an anti-seizure medication, is known to increase markers of oxidation and also increase the risk of neural tube defects (NTDs) when taken during pregnancy. It is unknown, however, whether oxidation plays a direct role in failed neural tube closure (NTC). Spatial and temporal fluctuations in total glutathione (GSH) and total cysteine (Cys) redox steady states were seen during a 24 h period of CD-1 mouse organogenesis in untreated conceptuses and following exposure to VPA and the Nrf2 antioxidant pathway inducer, 1,2-dithiole-3-thione (D3T). Glutathione, glutathione disulfide (GSSG), and Cys, cystine (CySS) concentrations, measured in conceptal tissues (embryo/visceral yolk sac) and fluids (yolk sac fluid/amniotic fluid) showed that VPA did not cause extensive and prolonged oxidation during the period of NTC, but instead produced transient periods of oxidation, as assessed by GSH:GSSG redox potentials, which revealed oxidation in all four conceptal compartments at 4, 10, and 14 h, corresponding to the period of heartbeat activation and NTC. Other changes were tissue and time specific. VPA treatment also reduced total FITC-Ab clearance from the medium over 3 h, indicating potential disruption of nutritive amino acid supply. Overall, these results indicated that VPA's ability to affect cellular redox status may be limited to tissue-specific windows of sensitivity during the period of NTC. The safety evaluation of drugs used during pregnancy should consider time and tissue specific redox factors.


Subject(s)
Anticonvulsants/toxicity , Antineoplastic Agents/toxicity , Embryo, Mammalian/drug effects , Thiones/toxicity , Thiophenes/toxicity , Valproic Acid/toxicity , Amino Acids/metabolism , Animals , Cysteine/metabolism , Embryo, Mammalian/metabolism , Female , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Maternal-Fetal Exchange , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Organogenesis/drug effects , Oxidation-Reduction , Pregnancy
18.
J Mol Med (Berl) ; 99(4): 489-500, 2021 04.
Article in English | MEDLINE | ID: mdl-33651139

ABSTRACT

Along with emergence of the organoids, their application in biomedical research has been currently one of the most fascinating themes. For the past few years, scientists have made significant contributions to deriving organoids representing the whole brain and specific brain regions. Coupled with somatic cell reprogramming and CRISPR/Cas9 editing, the organoid technologies were applied for disease modeling and drug screening. The methods to develop organoids further improved for rapid and efficient generation of cerebral organoids. Additionally, refining the methods to develop the regionally specified brain organoids enabled the investigation of development and interaction of the specific brain regions. Recent studies started resolving the issue in the lack of non-neuroectodermal cells in brain organoids, including vascular endothelial cells and microglia, which play fundamental roles in neurodevelopment and are involved in the pathophysiology of acute and chronic neural disorders. In this review, we highlight recent advances of neuronal organoid technologies, focusing on the region-specific brain organoids and complementation with endothelial cells and microglia, and discuss their potential applications to neuronal diseases.


Subject(s)
Brain/cytology , Endothelial Cells/cytology , Microglia/cytology , Organoids/cytology , Animals , Brain/anatomy & histology , Brain/embryology , Cellular Reprogramming Techniques/ethics , Cellular Reprogramming Techniques/methods , Embryoid Bodies/cytology , Fetus/anatomy & histology , Fibroblast Growth Factor 2/pharmacology , Hedgehog Proteins/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Neurons/cytology , Organ Specificity , Organogenesis/drug effects , Organoids/blood supply , Retina/cytology , Retina/embryology , Spinal Cord/cytology , Spinal Cord/embryology , Wnt Signaling Pathway
19.
J Cell Mol Med ; 25(3): 1350-1358, 2021 02.
Article in English | MEDLINE | ID: mdl-33393222

ABSTRACT

The hair follicle (HF) is an important mini-organ of the skin, composed of many types of cells. Dermal papilla cells are important signalling components that guide the proliferation, upward migration and differentiation of HF stem cell progenitor cells to form other types of HF cells. Thymosin ß4 (Tß4), a major actin-sequestering protein, is involved in various cellular responses and has recently been shown to play key roles in HF growth and development. Endogenous Tß4 can activate the mouse HF cycle transition and affect HF growth and development by promoting the migration and differentiation of HF stem cells and their progeny. In addition, exogenous Tß4 increases the rate of hair growth in mice and promotes cashmere production by increasing the number of secondary HFs (hair follicles) in cashmere goats. However, the molecular mechanisms through which Tß4 promotes HF growth and development have rarely been reported. Herein, we review the functions and mechanisms of Tß4 in HF growth and development and describe the endogenous and exogenous actions of Tß4 in HFs to provide insights into the roles of Tß4 in HF growth and development.


Subject(s)
Hair Follicle/cytology , Hair Follicle/physiology , Organogenesis , Thymosin/genetics , Thymosin/metabolism , Animals , Gene Expression Regulation/drug effects , Growth and Development/drug effects , Growth and Development/genetics , Hair Follicle/drug effects , Humans , Organogenesis/drug effects , Signal Transduction , Structure-Activity Relationship , Thymosin/chemistry , Thymosin/pharmacology
20.
Methods Mol Biol ; 2240: 103-117, 2021.
Article in English | MEDLINE | ID: mdl-33423230

ABSTRACT

The developing uterus is highly sensitive to a brief exposure to different substances, in particular those with endocrine-disrupting activity. Thus, exposure to environmental, nutritional, chemical, and other xenobiotic factors affecting signaling events during critical organizational periods can alter the normal course of uterine development with lasting consequences. In this chapter, we provide an experimental protocol to evaluate the development of the rat uterus as a toxicity biomarker at two different developmental time points: (1) the neonatal period, on postnatal day (PND) 8, and (2) the prepubertal period, on PND21. In this experimental approach, we propose to assess: (1) uterine morphology and cytodifferentiation, (2) uterine cell proliferation, and (3) the expression of proteins involved in uterine organogenetic differentiation. All these morphological and molecular markers are useful tools to determine the consequences of exposure to toxicants with the potential to disrupt the uterine development.


Subject(s)
Toxicity Tests , Uterus/drug effects , Age Factors , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Female , Immunohistochemistry , Microscopy , Organogenesis/drug effects , Pregnancy , Rats , Rats, Wistar , Uterus/metabolism , Uterus/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...