Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Publication year range
1.
Differentiation ; 135: 100743, 2024.
Article in English | MEDLINE | ID: mdl-38147763

ABSTRACT

The fovea centralis (fovea) is a specialized region of the primate retina that plays crucial roles in high-resolution visual acuity and color perception. The fovea is characterized by a high density of cone photoreceptors and no rods, and unique anatomical properties that contribute to its remarkable visual capabilities. Early histological analyses identified some of the key events that contribute to foveal development, but the mechanisms that direct the specification of this area are not understood. Recently, the expression of the retinoic acid-metabolizing enzyme CYP26A1 has become a hallmark of some of the retinal specializations found in vertebrates, including the primate fovea and the high-acuity area in avian species. In chickens, the retinoic acid pathway regulates the expression of FGF8 to then direct the development of a rod-free area. Similarly, high levels of CYP26A1, CDKN1A, and NPVF expression have been observed in the primate macula using transcriptomic approaches. However, which retinal cells express these genes and their expression dynamics in the developing primate eye remain unknown. Here, we systematically characterize the expression patterns of CYP26A1, FGF8, CDKN1A, and NPVF during the development of the rhesus monkey retina, from early stages of development in the first trimester until the third trimester (near term). Our data suggest that some of the markers previously proposed to be fovea-specific are not enriched in the progenitors of the rhesus monkey fovea. In contrast, CYP26A1 is expressed at high levels in the progenitors of the fovea, while it localizes in a subpopulation of macular Müller glia cells later in development. Together these data provide invaluable insights into the expression dynamics of several molecules in the nonhuman primate retina and highlight the developmental advancement of the foveal region.


Subject(s)
Chickens , Retina , Animals , Macaca mulatta/genetics , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Retinal Cone Photoreceptor Cells , Tretinoin
2.
Dev Dyn ; 253(4): 404-422, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37850839

ABSTRACT

BACKGROUND: Elongation of the spinal cord is dependent on neural development from neuromesodermal progenitors in the tail bud. We previously showed the involvement of the Oct4-type gene, pou5f3, in this process in zebrafish mainly by dominant-interference gene induction, but, to compensate for the limitation of this transgene approach, mutant analysis was indispensable. pou5f3 involvement in the signaling pathways was another unsolved question. RESULTS: We examined the phenotypes of pou5f3 mutants and the effects of Pou5f3 activation by the tamoxifen-ERT2 system in the posterior neural tube, together confirming the involvement of pou5f3. The reporter assays using P19 cells implicated tail bud-related transcription factors in pou5f3 expression. Regulation of tail bud development by retinoic acid (RA) signaling was confirmed by treatment of embryos with RA and the synthesis inhibitor, and in vitro reporter assays further showed that RA signaling regulated pou5f3 expression. Importantly, the expression of the RA degradation enzyme gene, cyp26a1, was down-regulated in embryos with disrupted pou5f3 activity. CONCLUSIONS: The involvement of pou5f3 in spinal cord extension was supported by using mutants and the gain-of-function approach. Our findings further suggest that pou5f3 regulates the RA level, contributing to neurogenesis in the posterior neural tube.


Subject(s)
Transcription Factors , Zebrafish , Animals , Gene Expression Regulation, Developmental , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Spinal Cord/metabolism , Transcription Factors/metabolism , Tretinoin/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
J Biol Chem ; 299(5): 104669, 2023 05.
Article in English | MEDLINE | ID: mdl-37011860

ABSTRACT

Considerable evidence confirms the importance of Cyp26a1 to all-trans-retinoic acid (RA) homeostasis during embryogenesis. In contrast, despite its presence in postnatal liver as a potential major RA catabolizing enzyme and its acute sensitivity to induction by RA, some data suggested that Cyp26a1 contributes only marginally to endogenous RA homeostasis postnatally. We report reevaluation of a conditional Cyp26a1 knockdown in the postnatal mouse. The current results show that Cyp26a1 mRNA in WT mouse liver increases 16-fold upon refeeding after a fast, accompanied by an increased rate of RA elimination and a 41% decrease in the RA concentration. In contrast, Cyp26a1 mRNA in the refed homozygotic knockdown reached only 2% of its extent in WT during refeeding, accompanied by a slower rate of RA catabolism and no decrease in liver RA, relative to fasting. Refed homozygous knockdown mice also had decreased Akt1 and 2 phosphorylation and pyruvate dehydrogenase kinase 4 (Pdk4) mRNA and increased glucokinase (Gck) mRNA, glycogen phosphorylase (Pygl) phosphorylation, and serum glucose, relative to WT. Fasted homozygous knockdown mice had increased glucagon/insulin relative to WT. These data indicate that Cyp26a1 participates prominently in moderating the postnatal liver concentration of endogenous RA and contributes essentially to glucoregulatory control.


Subject(s)
Blood Glucose , Homeostasis , Retinoic Acid 4-Hydroxylase , Tretinoin , Animals , Mice , Liver/enzymology , Liver/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , RNA, Messenger/genetics , Tretinoin/metabolism , Glucokinase/metabolism , Glycogen Phosphorylase/metabolism , Insulin/metabolism , Animals, Newborn , Phosphorylation , Blood Glucose/metabolism
4.
Dev Biol ; 486: 81-95, 2022 06.
Article in English | MEDLINE | ID: mdl-35364055

ABSTRACT

Endothelial cells (ECs) are critical to proper heart valve development, directly contributing to the mesenchyme of the cardiac cushions, which progressively transform into mature valves. To date, investigators have lacked sufficient markers of valve ECs to evaluate their contributions during valve morphogenesis fully. As a result, it has been unclear whether the well-characterized regional differentiation of valves correlates with any endothelial domains in the heart. Furthermore, it has been difficult to ascertain whether endothelial heterogeneity in the heart influences underlying mesenchymal zones in an angiocrine manner. To identify regionally expressed EC genes in the heart valves, we screened publicly available databases and assembled a toolkit of endothelial-enriched genes. We identified Cyp26b1 as one of many endothelial enriched genes found to be expressed in the endocardium of the developing cushions and valves. Here, we show that Cyp26b1 is required for normal heart valve development. Genetic ablation of Cyp26b1 in mouse embryos leads to abnormally thickened aortic valve leaflets, which is due in part to increased endothelial and mesenchymal cell proliferation in the remodeling valves. In addition, Cyp26b1 mutant hearts display ventricular septal defects (VSDs) in a portion of null embryos. We show that loss of Cyp26b1 results in upregulation of retinoic acid (RA) target genes, supporting the observation that Cyp26b1 has RA-dependent roles. Together, this work identifies a novel role for Cyp26b1 in heart valve morphogenesis and points to a role of RA in this process. Understanding the spatiotemporal expression dynamics of cardiac EC genes will pave the way for investigation of both normal and dysfunctional heart valve development.


Subject(s)
Endothelial Cells , Heart Valves , Animals , Aortic Valve , Heart Valves/metabolism , Mice , Morphogenesis , Organogenesis , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Tretinoin/metabolism
5.
Hum Genet ; 142(11): 1571-1586, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37755482

ABSTRACT

CYP26B1 metabolizes retinoic acid in the developing embryo to regulate its levels. A limited number of individuals with pathogenic variants in CYP26B1 have been documented with a varied phenotypic spectrum, spanning from a severe manifestation involving skull anomalies, craniosynostosis, encephalocele, radio-humeral fusion, oligodactyly, and a narrow thorax, to a milder presentation characterized by craniosynostosis, restricted radio-humeral joint mobility, hearing loss, and intellectual disability. Here, we report two families with CYP26B1-related phenotypes and describe the data obtained from functional studies of the variants. Exome and Sanger sequencing were used for variant identification in family 1 and family 2, respectively. Family 1 reflects a mild phenotype, which includes craniofacial dysmorphism with brachycephaly (without craniosynostosis), arachnodactyly, reduced radioulnar joint movement, conductive hearing loss, learning disability-and compound heterozygous CYP26B1 variants: (p.[(Pro118Leu)];[(Arg234Gln)]) were found. In family 2, a stillborn fetus presented a lethal phenotype with spina bifida occulta, hydrocephalus, poor skeletal mineralization, synostosis, limb defects, and a synonymous homozygous variant in CYP26B1: c.1083C > A. A minigene assay revealed that the synonymous variant created a new splice site, removing part of exon 5 (p.Val361_Asp382del). Enzymatic activity was assessed using a luciferase assay, demonstrating a notable reduction in exogenous retinoic acid metabolism for the variant p.Val361_Asp382del. (~ 3.5 × decrease compared to wild-type); comparatively, the variants p.(Pro118Leu) and p.(Arg234Gln) demonstrated a partial loss of metabolism (1.7× and 2.3× reduction, respectively). A proximity-dependent biotin identification assay reaffirmed previously reported ER-resident protein interactions. Additional work into these interactions is critical to determine if CYP26B1 is involved with other biological events on the ER. Immunofluorescence assay suggests that mutant CYP26B1 is still localized in the endoplasmic reticulum. These results indicate that novel pathogenic variants in CYP26B1 result in varying levels of enzymatic activity that impact retinoic acid metabolism and relate to the distinct phenotypes observed.


Subject(s)
Craniosynostoses , Tretinoin , Humans , Retinoic Acid 4-Hydroxylase/genetics , Tretinoin/metabolism , Homozygote , Exons , Craniosynostoses/genetics
6.
Development ; 147(15)2020 08 07.
Article in English | MEDLINE | ID: mdl-32665247

ABSTRACT

Retinoic acid (RA), a vitamin A (retinol) derivative, has pleiotropic functions during embryonic development. The synthesis of RA requires two enzymatic reactions: oxidation of retinol into retinaldehyde by alcohol dehydrogenases (ADHs) or retinol dehydrogenases (RDHs); and oxidation of retinaldehyde into RA by aldehyde dehydrogenases family 1, subfamily A (ALDH1as), such as ALDH1a1, ALDH1a2 and ALDH1a3. Levels of RA in tissues are regulated by spatiotemporal expression patterns of genes encoding RA-synthesizing and -degrading enzymes, such as cytochrome P450 26 (Cyp26 genes). Here, we show that RDH10 is important for both sensory and non-sensory formation of the vestibule of the inner ear. Mice deficient in Rdh10 exhibit failure of utricle-saccule separation, otoconial formation and zonal patterning of vestibular sensory organs. These phenotypes are similar to those of Aldh1a3 knockouts, and the sensory phenotype is complementary to that of Cyp26b1 knockouts. Together, these results demonstrate that RDH10 and ALDH1a3 are the key RA-synthesis enzymes involved in vestibular development. Furthermore, we discovered that RA induces Cyp26b1 expression in the developing vestibular sensory organs, which generates the differential RA signaling required for zonal patterning.


Subject(s)
Homeostasis , Organogenesis , Tretinoin/metabolism , Vestibule, Labyrinth/embryology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Mice , Mice, Knockout , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Vestibule, Labyrinth/cytology
7.
Development ; 147(4)2020 02 20.
Article in English | MEDLINE | ID: mdl-32001436

ABSTRACT

Proper organ development depends on coordinated communication between multiple cell types. Retinoic acid (RA) is an autocrine and paracrine signaling molecule essential for the development of most organs, including the lung. Despite extensive work detailing effects of RA deficiency in early lung morphogenesis, little is known about how RA regulates late gestational lung maturation. Here, we investigate the role of the RA catabolizing protein Cyp26b1 in the lung. Cyp26b1 is highly enriched in lung endothelial cells (ECs) throughout development. We find that loss of Cyp26b1 leads to reduction of alveolar type 1 cells, failure of alveolar inflation and early postnatal lethality in mouse. Furthermore, we observe expansion of distal epithelial progenitors, but no appreciable changes in proximal airways, ECs or stromal populations. Exogenous administration of RA during late gestation partially mimics these defects; however, transcriptional analyses comparing Cyp26b1-/- with RA-treated lungs reveal overlapping, but distinct, responses. These data suggest that defects observed in Cyp26b1-/- lungs are caused by both RA-dependent and RA-independent mechanisms. This work reports crucial cellular crosstalk during lung development involving Cyp26b1-expressing endothelium and identifies a novel RA modulator in lung development.


Subject(s)
Epithelium/embryology , Lung/embryology , Pulmonary Alveoli/embryology , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/physiology , Animals , CRISPR-Cas Systems , Cell Differentiation , Endothelial Cells/cytology , Epithelial Cells/cytology , Female , Gene Expression Regulation, Developmental , Kidney/embryology , Mice , Mice, Inbred C57BL , Organogenesis/drug effects , Pregnancy , Pregnancy, Animal , Signal Transduction , Stem Cells/cytology , Tretinoin/pharmacology
8.
Mol Carcinog ; 62(7): 991-1000, 2023 07.
Article in English | MEDLINE | ID: mdl-37042568

ABSTRACT

All-trans retinoic acid (ATRA) is the natural and synthetic analogue of vitamin A, playing an essential tumor suppressive role in multiple cancers including the esophageal squamous cell carcinoma (ESCC). Cytochrome P450 family 26 subfamily B member 1 (CYP26B1) exerts a critical regulator of ATRA levels through specific inactivation of ATRA to hydroxylated forms. Our previous exome-wide analyses revealed a rare missense variant in CYP26B1 significantly associated with ESCC risk in the Chinese population. However, it is still unclear whether there are common variants in CYP26B1 affect the susceptibility of ESCC and the tumor promotion role of CYP26B1 in vivo. In this research, we conducted a two-stage case-control study comprised of 5057 ESCC cases and 5397 controls, followed by a series of biochemical experiments to explore the function of CYP26B1 and its common variants in the tumorigenesis of ESCC. Intriguingly, we identified a missense variant rs2241057[A>G] in the fourth exon of CYP26B1 significantly associated with the ESCC risk (combined odds ratio = 1.28; 95% confidence interval = 1.15-1.42; p = 2.96 × 10-6 ). Through further functional analysis, we demonstrated that ESCC cells with the overexpression of rs2241057[G] had a significant lower level of retinoic acid, compared with the overexpression of rs2241057[A] or the control vector. In addition, the CYP26B1 overexpression and knock-out ESCC cells affected cell proliferation rate both in vitro and in vivo. These results highlighted the carcinogenicity of CYP26B1 related to the ATRA metabolism in ESCC risk.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/genetics , Retinoic Acid 4-Hydroxylase/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Case-Control Studies , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Tretinoin
9.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36769350

ABSTRACT

While genetic analyses have revealed ~100 risk loci associated with osteoarthritis (OA), only eight have been linked to hand OA. Besides, these studies were performed in predominantly European and Caucasian ancestries. Here, we conducted a genome-wide association study in the Han Chinese population to identify genetic variations associated with the disease. We recruited a total of 1136 individuals (n = 420 hand OA-affected; n = 716 unaffected control subjects) of Han Chinese ancestry. We carried out genotyping using Axiom Asia Precisi on Medicine Research Array, and we employed the RegulomeDB database and RoadMap DNase I Hypersensitivity Sites annotations to further narrow down our potential candidate variants. Genetic variants identified were tested in the Geisinger's hand OA cohort selected from the Geisinger MyCode community health initiative (MyCode®). We also performed a luciferase reporter assay to confirm the potential impact of top candidate single-nucleotide polymorphisms (SNPs) on hand OA. We identified six associated SNPs (p-value = 6.76 × 10-7-7.31 × 10-6) clustered at 2p13.2 downstream of the CYP26B1 gene. The strongest association signal identified was rs883313 (p-value = 6.76 × 10-7, odds ratio (OR) = 1.76), followed by rs12713768 (p-value = 1.36 × 10-6, OR = 1.74), near or within the enhancer region closest to the CYP26B1 gene. Our findings showed that the major risk-conferring CC haplotype of SNPs rs12713768 and rs10208040 [strong linkage disequilibrium (LD); D' = 1, r2 = 0.651] drives 18.9% of enhancer expression activity. Our findings highlight that the SNP rs12713768 is associated with susceptibility to and severity of hand OA in the Han Chinese population and that the suggested retinoic acid signaling pathway may play an important role in its pathogenesis.


Subject(s)
Osteoarthritis , Vitamin A , Humans , Retinoic Acid 4-Hydroxylase/genetics , Genome-Wide Association Study , Genetic Predisposition to Disease , Alleles , Osteoarthritis/genetics , Polymorphism, Single Nucleotide , Genes, Regulator , Case-Control Studies , Genotype , China
10.
Hum Mol Genet ; 29(2): 264-273, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31813960

ABSTRACT

Glycogen storage disease type 1a (GSD Ia) is an inborn error of metabolism caused by mutations in the G6PC gene, encoding the catalytic subunit of glucose-6-phosphatase. Early symptoms include severe fasting intolerance, failure to thrive and hepatomegaly, biochemically associated with nonketotic hypoglycemia, fasting hyperlactidemia, hyperuricemia and hyperlipidemia. Dietary management is the cornerstone of treatment aiming at maintaining euglycemia, prevention of secondary metabolic perturbations and long-term complications, including liver (hepatocellular adenomas and carcinomas), kidney and bone disease (hypovitaminosis D and osteoporosis). As impaired vitamin A homeostasis also associates with similar symptoms and is coordinated by the liver, we here analysed whether vitamin A metabolism is affected in GSD Ia patients and liver-specific G6pc-/- knock-out mice. Serum levels of retinol and retinol binding protein 4 (RBP4) were significantly increased in both GSD Ia patients and L-G6pc-/- mice. In contrast, hepatic retinol levels were significantly reduced in L-G6pc-/- mice, while hepatic retinyl palmitate (vitamin A storage form) and RBP4 levels were not altered. Transcript and protein analyses indicate an enhanced production of retinol and reduced conversion the retinoic acids (unchanged LRAT, Pnpla2/ATGL and Pnpla3 up, Cyp26a1 down) in L-G6pc-/- mice. Aberrant expression of genes involved in vitamin A metabolism was associated with reduced basal messenger RNA levels of markers of inflammation (Cd68, Tnfα, Nos2, Il-6) and fibrosis (Col1a1, Acta2, Tgfß, Timp1) in livers of L-G6pc-/- mice. In conclusion, GSD Ia is associated with elevated serum retinol and RBP4 levels, which may contribute to disease symptoms, including osteoporosis and hepatic steatosis.


Subject(s)
Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/metabolism , Liver/metabolism , Retinol-Binding Proteins, Plasma/metabolism , Vitamin A/blood , Adolescent , Adult , Animals , Diterpenes/metabolism , Fatty Liver/metabolism , Female , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/blood , Glycogen Storage Disease Type I/enzymology , Glycogen Storage Disease Type I/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Osteoporosis/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Retinol-Binding Proteins, Plasma/genetics , Retinyl Esters , Vitamin A/analogs & derivatives , Vitamin A/metabolism
11.
J Cell Mol Med ; 25(3): 1771-1782, 2021 02.
Article in English | MEDLINE | ID: mdl-33438367

ABSTRACT

Cyp26a1 had important roles in mouse embryo implantation and was highly expressed in some of NK cells at the human maternal-foetal interface in early pregnancy. However, the regulatory effect of Cyp26a1 on NK cells remains poorly understood. Through qPCR and flow cytometric assays, we found that Cyp26a1 was expressed by mouse uterine NK cells but not spleen NK cells during the peri-implantation period and there was a group of NK cells that highly expressed Cyp26a1, that is Cyp26a1+ NK cell subset. single cell-population transcriptome sequencing on Cyp26a1+ NK and Cyp26a1- NK cell subsets was performed. We found that there were 3957 differentially expressed genes in the Cyp26a1+ NK cell subset with a cut-off of fold change ≥2 and FDR < 0.01, 2509 genes were up-regulated and 1448 genes were down-regulated in Cyp26a1+ NK cell subset. Moreover, cytokine-cytokine receptor interaction signalling pathway and natural killer cell-mediated cytotoxicity signalling pathway were enriched according to KEGG pathway enrichment analysis. We further found that the expression of Gzma and Klrg1 was significantly increased and Fcgr4 was significantly decreased when inhibiting Cyp26a1. Our experimental results show that there is a novel NK cell subset of Cyp26a1+ NK cells in mouse uterus and Cyp26a1 can regulate the gene expression of Gzma, Klrg1 and Fcgr4 in the Cyp26a1+ NK cells.


Subject(s)
Gene Expression , Killer Cells, Natural/metabolism , Lymphocyte Subsets/metabolism , Placenta/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Animals , Computational Biology/methods , Female , Gene Expression Profiling , Immunohistochemistry , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Mice , Placenta/immunology , Pregnancy , Retinoic Acid 4-Hydroxylase/metabolism , Transcriptome
12.
Development ; 145(9)2018 05 08.
Article in English | MEDLINE | ID: mdl-29650589

ABSTRACT

Zebrafish mutants with increased retinoic acid (RA) signaling due to the loss of the RA-inactivating enzyme Cyp26b1 develop a hyper-mineralized spine with gradually fusing vertebral body precursors (centra). However, the underlying cellular mechanisms remain incompletely understood. Here, we show that cells of the notochord epithelium named chordoblasts are sensitive to RA signaling. Chordoblasts are uniformly distributed along the anteroposterior axis and initially generate the continuous collagenous notochord sheath. However, subsequently and iteratively, subsets of these cells undergo further RA-dependent differentiation steps, acquire a stellate-like shape, downregulate expression of the collagen gene col2a1a, switch on cyp26b1 expression and trigger metameric sheath mineralization. This mineralization fails to appear upon chordoblast-specific cell ablation or RA signal transduction blockade. Together, our data reveal that, despite their different developmental origins, the activities and regulation of chordoblasts are very similar to those of osteoblasts, including their RA-induced transition from osteoid-producing cells to osteoid-mineralizing ones. Furthermore, our data point to a requirement for locally controlled RA activity within the chordoblast layer in order to generate the segmented vertebral column.


Subject(s)
Calcification, Physiologic/physiology , Gene Expression Regulation, Developmental/physiology , Notochord/embryology , Spine/embryology , Tretinoin/metabolism , Zebrafish/embryology , Animals , Collagen/biosynthesis , Collagen/genetics , Notochord/cytology , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Spine/cytology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Development ; 145(18)2018 09 17.
Article in English | MEDLINE | ID: mdl-30111657

ABSTRACT

The developing vertebrate embryo is exquisitely sensitive to retinoic acid (RA) concentration, particularly during anteroposterior patterning. In contrast to Nodal and Wnt signaling, RA was not previously considered to be an instructive signal in mesoderm formation during gastrulation. Here, we show in Xenopus that RARγ is indispensable for the expression of early mesoderm markers and is, therefore, an obligatory factor in mesodermal competence and/or maintenance. We identified several novel targets upregulated by RA receptor signaling in the early gastrula that are expressed in the circumblastoporal ring and linked to mesodermal development. Despite overlapping expression patterns of the genes encoding the RA-synthesizing enzyme Aldh1a2 and the RA-degrading enzyme Cyp26a1, RARγ1 functions as a transcriptional activator in early mesoderm development, suggesting that RA ligand is available to the embryo earlier than previously appreciated. RARγ1 is required for cellular adhesion, as revealed by spontaneous dissociation and depletion of ncam1 mRNA in animal caps harvested from RARγ1 knockdown embryos. RARγ1 knockdown obliterates somite boundaries, and causes loss of Myod protein in the presomitic mesoderm, but ectopic, persistent expression of Myod protein in the trunk. Thus, RARγ1 is required for stabilizing the mesodermal fate, myogenic commitment, somite boundary formation, and terminal skeletal muscle differentiation.


Subject(s)
Body Patterning/genetics , Mesoderm/embryology , Muscle, Skeletal/embryology , Receptors, Retinoic Acid/genetics , Xenopus laevis/embryology , Aldehyde Dehydrogenase 1 Family , Aldehyde Oxidase/biosynthesis , Aldehyde Oxidase/genetics , Animals , CD56 Antigen/metabolism , Cell Adhesion/genetics , Gastrulation/genetics , MyoD Protein/metabolism , Receptors, Retinoic Acid/metabolism , Retinal Dehydrogenase , Retinoic Acid 4-Hydroxylase/biosynthesis , Retinoic Acid 4-Hydroxylase/genetics , Signal Transduction/genetics , Transcriptional Activation/genetics , Tretinoin/metabolism , Xenopus Proteins/biosynthesis , Xenopus Proteins/genetics , Xenopus laevis/genetics , Retinoic Acid Receptor gamma
14.
Toxicol Appl Pharmacol ; 433: 115781, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34737147

ABSTRACT

The cardiac embryonic stem cell test (ESTc) is an in vitro embryotoxicity screen which uses cardiomyocyte formation as the main differentiation route. Studies are ongoing into whether an improved specification of the biological domain can broaden the applicability of the test, e.g. to discriminate between structurally similar chemicals by measuring expression of dedicated gene transcript biomarkers. We explored this with two chemical classes: morpholines (tridemorph; fenpropimorph) and piperidines (fenpropidin; spiroxamine). These compounds cause embryotoxicity in rat such as cleft palate. This malformation can be linked to interference with retinoic acid balance, neural crest (NC) cell migration, or cholesterol biosynthesis. Also neural differentiation within the ESTc was explored in relation to these compounds. Gene transcript expression of related biomarkers were measured at low and high concentrations on differentiation day 4 (DD4) and DD10. All compounds showed stimulating effects on the cholesterol biosynthesis related marker Msmo1 after 24 h exposure and tridemorph showed inhibition of Cyp26a1 which codes for one of the enzymes that metabolises retinoic acid. A longer exposure duration enhanced expression levels for differentiation markers for cardiomyocytes (Nkx2-5; Myh6) and neural cells (Tubb3) on DD10. This readout gave additional mechanistic insight which enabled previously unavailable in vitro discrimination between the compounds, showing the practical utility of specifying the biological domain of the ESTc.


Subject(s)
Cell Differentiation/drug effects , Gene Expression Regulation, Developmental/drug effects , Morpholines/toxicity , Mouse Embryonic Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Piperidines/toxicity , Toxicity Tests , Animals , Cells, Cultured , Gene Regulatory Networks , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Mice , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Risk Assessment , Spiro Compounds/toxicity , Time Factors , Tubulin/genetics , Tubulin/metabolism
15.
Am J Med Genet A ; 185(9): 2766-2775, 2021 09.
Article in English | MEDLINE | ID: mdl-34160123

ABSTRACT

Retinoic acid exposures as well as defects in the retinoic acid-degrading enzyme CYP26B1 have teratogenic effects on both limb and craniofacial skeleton. An initial report of four individuals described a syndrome of fetal and infantile lethality with craniosynostosis and skeletal anomalies caused by homozygous pathogenic missense variants in CYP26B1. In contrast, a 22-year-old female was reported with a homozygous missense pathogenic variant in CYP26B1 with complex multisuture craniosynostosis and intellectual disability, suggesting that in some cases, biallelic pathogenic variants of CYP26B1 may be compatible with life. Here we describe four additional living individuals from two families with compound heterozygous pathogenic missense variants in CYP26B1. Structural assessment of these additional missense variants places them further from the catalytic site and supports a model consistent with milder nonlethal disease. In addition to previously reported findings of multisuture craniosynostosis, conductive hearing loss, joint contractures, long slender fingers, camptodactly, broad fingertips, and developmental delay/intellectual disability, skeletal imaging in our cases also revealed gracile long bones, gracile ribs, radioulnar synostosis, and carpal and/or tarsal fusions. These individuals broaden the phenotypic range of biallelic pathogenic variants in CYPB26B1 and most significantly clarify that mortality can range from perinatal lethality to survival into adulthood.


Subject(s)
Abnormalities, Multiple/pathology , Homozygote , Mutation, Missense , Radius/abnormalities , Retinoic Acid 4-Hydroxylase/genetics , Synostosis/pathology , Ulna/abnormalities , Abnormalities, Multiple/genetics , Child , Family , Female , Humans , Infant , Male , Phenotype , Radius/pathology , Synostosis/genetics , Ulna/pathology
16.
J Immunol ; 202(9): 2795-2805, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30885956

ABSTRACT

Recent studies have underscored the critical role of retinoic acid (RA) in the development of lineage-committed CD4 and CD8 T cells in vivo. We have shown that under acute graft-versus-host disease (GVHD) inflammatory conditions, RA is upregulated in the intestine and is proinflammatory, as GVHD lethality was attenuated when donor allogeneic T cells selectively expressed a dominant negative RA receptor α that blunted RA signaling. RA can function in an autocrine and paracrine fashion, and as such, the host cell lineage responsible for the production of RA metabolism and the specific RA-metabolizing enzymes that potentiate GVHD severity are unknown. In this study, we demonstrate that enhancing RA degradation in the host and to a lesser extent donor hematopoietic cells by overexpressing the RA-catabolizing enzyme CYP26A1 reduced GVHD. RA production is facilitated by retinaldehyde isoform-2 (RALDH2) preferentially expressed in dendritic cells (DCs). Conditionally deleted RA-synthesizing enzyme RALDH2 in host or to a lesser extent donor DCs reduced GVHD lethality. Improved survival in recipients with RALDH2-deleted DCs was associated with increased T cell death, impaired T effector function, increased regulatory T cell frequency, and augmented coinhibitory molecule expression on donor CD4+ T cells. In contrast, retinaldehydrogenase isoform-1 (RALDH1) is dominantly expressed in intestinal epithelial cells. Unexpectedly, conditional host intestinal epithelial cells RALDH1 deletion failed to reduce GVHD. These data demonstrate the critical role of both donor and especially host RALDH2+ DCs in driving murine GVHD and suggest RALDH2 inhibition or CYP26A1 induction as novel therapeutic strategies to prevent GVHD.


Subject(s)
Aldehyde Oxidoreductases/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Expression Regulation, Enzymologic/immunology , Graft vs Host Disease/immunology , Aldehyde Oxidoreductases/genetics , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/immunology , Tretinoin/immunology
17.
Biotechnol Lett ; 43(2): 415-422, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33179169

ABSTRACT

The comparative transcriptome analysis of the fungus Gibberella zeae which could efficiently catalyze the 7ß-hydroxylation of LCA to produce UDCA was performed with LCA induction. This is the first time to report the comparative transcriptome of fungus under LCA treatment. Totally, 1364 differentially expressed genes including 770 up-regulated and 594 down-regulated genes were identified. In the 770 up-regulated genes, 12 genes with the function of hydroxylation were picked out by application of function screening, which were annotated as CYP450 or hydroxylase. Moreover, the qRT-PCR results of five up-regulated CYP450-like genes confirmed the credibility of RNA-Seq further. These results provide valuable information for the discovery of novel enzyme producing clinical drug UDCA from butchery byproduct LCA, and also might indicate some clues for the detoxification process of LCA in humans.


Subject(s)
Fusarium/genetics , Lithocholic Acid/metabolism , Transcriptome/genetics , Ursodeoxycholic Acid/metabolism , Gene Expression Regulation, Fungal/genetics , Humans , Lithocholic Acid/genetics , Mixed Function Oxygenases/genetics , Retinoic Acid 4-Hydroxylase/genetics , Ursodeoxycholic Acid/genetics
18.
J Biol Chem ; 294(29): 11166-11179, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31167781

ABSTRACT

The all-trans-retinoic acid (atRA) hydroxylase Cyp26a1 is essential for embryonic development and may play a key role in regulating atRA clearance also in adults. We hypothesized that loss of Cyp26a1 activity via inducible knockout in juvenile or adult mice would result in decreased atRA clearance and increased tissue atRA concentrations and atRA-related adverse effects. To test these hypotheses, Cyp26a1 was knocked out in juvenile and adult male and female Cyp26a1 floxed mice using standard Cre-Lox technology and tamoxifen injections. Biochemical and histological methods were used to study the effects of global Cyp26a1 knockout. The Cyp26a1 knockout did not result in consistent histopathological changes in any major organs. Cyp26a1-/- mice gained weight normally and exhibited no adverse phenotypes for up to 1 year after loss of Cyp26a1 expression. Similarly, atRA concentrations were not increased in the liver, testes, spleen, or serum of these mice, and the Cyp26a1 knockout did not cause compensatory induction of lecithin:retinol acetyltransferase (Lrat) or retinol dehydrogenase 11 (Rdh11) mRNA or a decrease in aldehyde dehydrogenase 1a1 (Aldh1a1) mRNA in the liver compared with tamoxifen-treated controls. However, the Cyp26a1-/- mice showed increased bone marrow cellularity and decreased frequency of erythroid progenitor cells in the bone marrow consistent with a retinoid-induced myeloid skewing of hematopoiesis. In addition, the Cyp26a1 knockout decreased clearance of exogenous atRA by 70% and increased atRA half-life 6-fold. These findings demonstrate that despite lacking a major impact on endogenous atRA signaling, Cyp26a1 critically contributes as a barrier for exogenous atRA exposure.


Subject(s)
Homeostasis , Retinoic Acid 4-Hydroxylase/metabolism , Tretinoin/pharmacokinetics , Vitamin A/metabolism , Acyltransferases/genetics , Aldehyde Dehydrogenase 1 Family/genetics , Animals , Mice , Mice, Knockout , Oxidoreductases/genetics , RNA, Messenger/genetics , Retinal Dehydrogenase/genetics , Retinoic Acid 4-Hydroxylase/genetics , Signal Transduction , Tamoxifen/administration & dosage
19.
J Biol Chem ; 294(14): 5536-5548, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30737277

ABSTRACT

Ethanol (EtOH) is a teratogen, but its teratogenic mechanisms are not fully understood. The alcohol form of vitamin A (retinol/ROL) can be oxidized to all-trans-retinoic acid (RA), which plays a critical role in stem cell differentiation and development. Using an embryonic stem cell (ESC) model to analyze EtOH's effects on differentiation, we show here that EtOH and acetaldehyde, but not acetate, increase differentiation-associated mRNA levels, and that EtOH decreases pluripotency-related mRNAs. Using reporter assays, ChIP assays, and retinoic acid receptor-γ (RARγ) knockout ESC lines generated by CRISPR/Cas9 and homologous recombination, we demonstrate that EtOH signals via RARγ binding to RA response elements (RAREs) in differentiation-associated gene promoters or enhancers. We also report that EtOH-mediated increases in homeobox A1 (Hoxa1) and cytochrome P450 family 26 subfamily A member 1 (Cyp26a1) transcripts, direct RA target genes, require the expression of the RA-synthesizing enzyme, aldehyde dehydrogenase 1 family member A2 (Aldh1a2), suggesting that EtOH-mediated induction of Hoxa1 and Cyp26a1 requires ROL from the serum. As shown with CRISPR/Cas9 knockout lines, the retinol dehydrogenase gene Rdh10 and a functional RARE in the ROL transporter stimulated by retinoic acid 6 (Stra6) gene are required for EtOH induction of Hoxa1 and Cyp26a1 We conclude that EtOH stimulates stem cell differentiation by increasing the influx and metabolism of ROL for downstream RARγ-dependent transcription. In stem cells, EtOH may shift cell fate decisions to alter developmental outcomes by increasing endogenous ROL/RA signaling via increased Stra6 expression and ROL oxidation.


Subject(s)
Cell Differentiation/drug effects , Ethanol/pharmacology , Mouse Embryonic Stem Cells/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction/drug effects , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Cell Differentiation/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Receptors, Retinoic Acid/genetics , Retinal Dehydrogenase , Retinoic Acid 4-Hydroxylase/biosynthesis , Retinoic Acid 4-Hydroxylase/genetics , Signal Transduction/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Retinoic Acid Receptor gamma
20.
J Exp Zool B Mol Dev Evol ; 334(3): 156-167, 2020 05.
Article in English | MEDLINE | ID: mdl-32037722

ABSTRACT

The body color of the Pleuronectiformes is bilaterally asymmetric between right and left halves, with a dark ocular-side and a white blind-side. This body color asymmetry develops by restricted differentiation of melanophores and xanthophores on the ocular-side during metamorphosis, accompanied by migration of one eye to the future ocular-side. In this study, we elucidated the developmental regulatory system of this lateralized pigmentation. We found that in flounder, Sox10-positive chromatophore progenitors appear bilaterally both in the ocular- and blind-side skin of metamorphosing larvae, and that those arriving at the ocular-side skin differentiate into gch2-positive chromatoblasts and then chromatophores. Transient exposure of metamorphosing larvae to retinoic acid (RA)-induced progenitors on the blind-side to differentiate into gch2-positive chromatoblasts. On the contrary, exposure to an RA receptor antagonist, BMS493, suppressed the differentiation of gch2-positive chromatoblasts on the ocular-side. Thus, we demonstrated that RA is essential for flounder chromatophore progenitors to differentiate into chromatoblasts. At the time of chromatoblast differentiation on the ocular-side, cyp26b1, which inactivates RA, was upregulated on the blind-side skin compared with the ocular-side. Therefore, we surmise that ocular-side-specific pigmentation is regulated by the inhibition of RA-signaling by cyp26b1 on the blind-side.


Subject(s)
Flounder/physiology , Pigmentation/physiology , Tretinoin/metabolism , Animals , Benzoates/pharmacology , Gene Expression Regulation/physiology , Larva/physiology , Metamorphosis, Biological/physiology , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Signal Transduction , Skin/metabolism , Stilbenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL