Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.015
Filter
1.
Cells ; 13(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39273043

ABSTRACT

A complete understanding of neural crest cell mechanodynamics during ocular development will provide insight into postnatal neural crest cell contributions to ophthalmic abnormalities in adult tissues and inform regenerative strategies toward injury repair. Herein, single-cell RNA sequencing in zebrafish during early eye development revealed keratin intermediate filament genes krt8 and krt18a.1 as additional factors expressed during anterior segment development. In situ hybridization and immunofluorescence microscopy confirmed krt8 and krt18a.1 expression in the early neural plate border and migrating cranial neural crest cells. Morpholino oligonucleotide (MO)-mediated knockdown of K8 and K18a.1 markedly disrupted the migration of neural crest cell subpopulations and decreased neural crest cell marker gene expression in the craniofacial region and eye at 48 h postfertilization (hpf), resulting in severe phenotypic defects reminiscent of neurocristopathies. Interestingly, the expression of K18a.1, but not K8, is regulated by retinoic acid (RA) during early-stage development. Further, both keratin proteins were detected during postnatal corneal regeneration in adult zebrafish. Altogether, we demonstrated that both K8 and K18a.1 contribute to the early development and postnatal repair of neural crest cell-derived ocular tissues.


Subject(s)
Cornea , Keratin-8 , Neural Crest , Regeneration , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Neural Crest/metabolism , Neural Crest/cytology , Keratin-8/metabolism , Keratin-8/genetics , Cornea/metabolism , Gene Expression Regulation, Developmental/drug effects , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Keratin-18/metabolism , Keratin-18/genetics , Tretinoin/pharmacology , Tretinoin/metabolism , Cell Movement/genetics
2.
J Cell Mol Med ; 28(15): e18584, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135338

ABSTRACT

Breast cancer (BC) is still one of the major issues in world health, especially for women, which necessitates innovative therapeutic strategies. In this study, we investigated the efficacy of retinoic acid derivatives as inhibitors of 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1), which plays a crucial role in the biosynthesis and metabolism of oestrogen and thereby influences the progression of BC and, the main objective of this investigation is to identify the possible drug candidate against BC through computational drug design approach including PASS prediction, molecular docking, ADMET profiling, molecular dynamics simulations (MD) and density functional theory (DFT) calculations. The result has reported that total eight derivatives with high binding affinity and promising pharmacokinetic properties among 115 derivatives. In particular, ligands 04 and 07 exhibited a higher binding affinity with values of -9.9 kcal/mol and -9.1 kcal/mol, respectively, than the standard drug epirubicin hydrochloride, which had a binding affinity of -8.2 kcal/mol. The stability of the ligand-protein complexes was further confirmed by MD simulations over a 100-ns trajectory, which included assessments of hydrogen bonds, root mean square deviation (RMSD), root mean square Fluctuation (RMSF), dynamic cross-correlation matric (DCCM) and principal component analysis. The study emphasizes the need for experimental validation to confirm the therapeutic utility of these compounds. This study enhances the computational search for new BC drugs and establishes a solid foundation for subsequent experimental and clinical research.


Subject(s)
Breast Neoplasms , Molecular Docking Simulation , Molecular Dynamics Simulation , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Female , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Ligands , Computer Simulation , Protein Binding , Tretinoin/metabolism , Drug Design , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , Hydrogen Bonding
3.
Nat Commun ; 15(1): 6852, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127768

ABSTRACT

Cis-regulatory elements (CREs) are pivotal in orchestrating gene expression throughout diverse biological systems. Accurate identification and in-depth characterization of functional CREs are crucial for decoding gene regulation networks during cellular processes. In this study, we develop Kethoxal-Assisted Single-stranded DNA Assay for Transposase-Accessible Chromatin with Sequencing (KAS-ATAC-seq) to quantitatively analyze the transcriptional activity of CREs. A main advantage of KAS-ATAC-seq lies in its precise measurement of ssDNA levels within both proximal and distal ATAC-seq peaks, enabling the identification of transcriptional regulatory sequences. This feature is particularly adept at defining Single-Stranded Transcribing Enhancers (SSTEs). SSTEs are highly enriched with nascent RNAs and specific transcription factors (TFs) binding sites that define cellular identity. Moreover, KAS-ATAC-seq provides a detailed characterization and functional implications of various SSTE subtypes. Our analysis of CREs during mouse neural differentiation demonstrates that KAS-ATAC-seq can effectively identify immediate-early activated CREs in response to retinoic acid (RA) treatment. Our findings indicate that KAS-ATAC-seq provides more precise annotation of functional CREs in transcription. Future applications of KAS-ATAC-seq would help elucidate the intricate dynamics of gene regulation in diverse biological processes.


Subject(s)
Transcription Factors , Animals , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic , Enhancer Elements, Genetic/genetics , Chromatin/metabolism , Chromatin/genetics , Binding Sites , Humans , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Chromatin Immunoprecipitation Sequencing/methods , Transposases/metabolism , Transposases/genetics , Regulatory Elements, Transcriptional , Tretinoin/pharmacology , Tretinoin/metabolism , Gene Expression Regulation , Cell Differentiation/genetics , Sequence Analysis, DNA/methods , Regulatory Sequences, Nucleic Acid/genetics
4.
Genome Biol ; 25(1): 211, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118163

ABSTRACT

BACKGROUND: The Pharyngeal Endoderm (PE) is an extremely relevant developmental tissue, serving as the progenitor for the esophagus, parathyroids, thyroids, lungs, and thymus. While several studies have highlighted the importance of PE cells, a detailed transcriptional and epigenetic characterization of this important developmental stage is still missing, especially in humans, due to technical and ethical constraints pertaining to its early formation. RESULTS: Here we fill this knowledge gap by developing an in vitro protocol for the derivation of PE-like cells from human Embryonic Stem Cells (hESCs) and by providing an integrated multi-omics characterization. Our PE-like cells robustly express PE markers and are transcriptionally homogenous and similar to in vivo mouse PE cells. In addition, we define their epigenetic landscape and dynamic changes in response to Retinoic Acid by combining ATAC-Seq and ChIP-Seq of histone modifications. The integration of multiple high-throughput datasets leads to the identification of new putative regulatory regions and to the inference of a Retinoic Acid-centered transcription factor network orchestrating the development of PE-like cells. CONCLUSIONS: By combining hESCs differentiation with computational genomics, our work reveals the epigenetic dynamics that occur during human PE differentiation, providing a solid resource and foundation for research focused on the development of PE derivatives and the modeling of their developmental defects in genetic syndromes.


Subject(s)
Cell Differentiation , Endoderm , Epigenesis, Genetic , Human Embryonic Stem Cells , Humans , Endoderm/cytology , Endoderm/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Pharynx/cytology , Pharynx/metabolism , Tretinoin/pharmacology , Tretinoin/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Transcription Factors/genetics , Mice
5.
Toxicology ; 508: 153907, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39121937

ABSTRACT

Gut microbiota symbiosis faces enormous challenge with increasing exposure to drugs such as environmental poisons and antibiotics. The gut microbiota is an important component of the host microbiota and has been proven to be involved in regulating spermatogenesis, but the molecular mechanism is still unclear. A male mouse model with gut microbiota depletion/dysbiosis was constructed by adding combined antibiotics to free drinking water, and reproductive parameters such as epididymal sperm count, testicular weight and paraffin sections were measured. Testicular transcriptomic and serum metabolomic analyses were performed to reveal the molecular mechanism of reproductive dysfunction induced by gut microbiota dysbiosis in male mice.This study confirms that antibiotic induced depletion of gut microbiota reduces sperm count in the epididymis and reduces germ cells in the seminiferous tubules in male mice. Further study showed that exosomes isolated from microbiota-depleted mice led to abnormally high levels of retinoic acid and decrease in the number of germ cells in the seminiferous tubules and sperm in the epididymis. Finally, abnormally high levels of retinoic acid was confirmed to disrupted meiotic processes, resulting in spermatogenesis disorders. This study proposed the concept of the gut microbiota-exosome-retinoic acid-testicular axis and demonstrated that depletion of the gut microbiota caused changes in the function of exosomes, which led to abnormal retinoic acid metabolism in the testis, thereby impairing meiosis and spermatogenesis processes.


Subject(s)
Dysbiosis , Exosomes , Gastrointestinal Microbiome , Spermatogenesis , Testis , Tretinoin , Animals , Male , Spermatogenesis/drug effects , Tretinoin/metabolism , Gastrointestinal Microbiome/drug effects , Exosomes/metabolism , Exosomes/drug effects , Mice , Testis/drug effects , Testis/metabolism , Testis/pathology , Dysbiosis/chemically induced , Anti-Bacterial Agents/toxicity , Mice, Inbred C57BL , Epididymis/drug effects , Epididymis/metabolism , Epididymis/pathology , Sperm Count , Spermatozoa/drug effects , Spermatozoa/metabolism , Spermatozoa/pathology
6.
Int J Mol Sci ; 25(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39201764

ABSTRACT

Fish retinal ganglion cells (RGCs) can regenerate after optic nerve lesions (ONLs). We previously reported that heat shock factor 1 (HSF1) and Yamanaka factors increased in the zebrafish retina 0.5-24 h after ONLs, and they led to cell survival and the transformation of neuro-stem cells. We also showed that retinoic acid (RA) signaling and transglutaminase 2 (TG2) were activated in the fish retina, performing neurite outgrowth 5-30 days after ONLs. In this study, we found that RA signaling and TG2 increased within 0.5 h in the zebrafish retina after ONLs. We examined their interaction with the TG2-specific morpholino and inhibitor due to the significantly close initiation time of TG2 and HSF1. The inhibition of TG2 led to the complete suppression of HSF1 expression. Furthermore, the results of a ChIP assay with an anti-TG2 antibody evidenced significant anti-TG2 immunoprecipitation of HSF1 genome DNA after ONLs. The inhibition of TG2 also suppressed Yamanaka factors' gene expression. This rapid increase in TG2 expression occurred 30 min after the ONLs, and RA signaling occurred 15 min before this change. The present study demonstrates that TG2 regulates Yamanaka factors via HSF1 signals in the acute phase of fish optic nerve regeneration.


Subject(s)
Heat Shock Transcription Factors , Nerve Regeneration , Optic Nerve , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases , Zebrafish , Animals , Zebrafish/genetics , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism , Nerve Regeneration/genetics , Optic Nerve/metabolism , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Tretinoin/pharmacology , Tretinoin/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Retinal Ganglion Cells/metabolism , Gene Expression Regulation/drug effects , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/genetics , Signal Transduction
7.
Stem Cell Reports ; 19(9): 1336-1350, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39151428

ABSTRACT

Variability between human pluripotent stem cell (hPSC) lines remains a challenge and opportunity in biomedicine. In this study, hPSC lines from multiple donors were differentiated toward neuroectoderm and mesendoderm lineages. We revealed dynamic transcriptomic patterns that delineate the emergence of these lineages, which were conserved across lines, along with individual line-specific transcriptional signatures that were invariant throughout differentiation. These transcriptomic signatures predicted an antagonism between SOX21-driven forebrain fates and retinoic acid-induced hindbrain fates. Replicate lines and paired adult tissue demonstrated the stability of these line-specific transcriptomic traits. We show that this transcriptomic variation in lineage bias had both genetic and epigenetic origins, aligned with the anterior-to-posterior structure of early mammalian development, and was present across a large collection of hPSC lines. These findings contribute to developing systematic analyses of PSCs to define the origin and consequences of variation in the early events orchestrating individual human development.


Subject(s)
Cell Differentiation , Cell Lineage , Pluripotent Stem Cells , Transcriptome , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Line , Tretinoin/pharmacology , Tretinoin/metabolism , Gene Expression Regulation, Developmental , Epigenesis, Genetic
8.
J Immunol ; 213(7): 933-939, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39132993

ABSTRACT

The most common congenital viral infection is CMV, which leads to numerous neurologic disabilities. Using a mouse model of congenital CMV, we previously determined that Ag-specific CD8+ T cells traffic to the brain in a CCR9-dependent manner. The mechanism by which these CD8+ T cells acquire a CCR9-dependent "brain-tropic" phenotype remains unclear. In this study, we identify the key factor that imprints brain homing specificity on CD8+ T cells, the source of production, and the location where CCR9 expression is induced. Specifically, we discovered that CCR9 is induced on CD8+ T cells by retinoic acid-producing CD8α+ dendritic cells in the cervical lymph node postinfection. We found that retinoic acid is important for CD8+ T cells to establish tissue residency in the brain. Collectively, our data expand the role of retinoic acid during infection and mechanistically demonstrate how CD8+ T cells are primed to protect the brain during congenital viral infection.


Subject(s)
Brain , CD8-Positive T-Lymphocytes , Cytomegalovirus Infections , Tretinoin , Animals , CD8-Positive T-Lymphocytes/immunology , Tretinoin/metabolism , Mice , Cytomegalovirus Infections/immunology , Brain/immunology , Mice, Inbred C57BL , Dendritic Cells/immunology , Cytomegalovirus/immunology , Disease Models, Animal , Cell Movement/immunology
9.
Sci Rep ; 14(1): 20222, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39215116

ABSTRACT

The adult mammalian heart has extremely limited cardiac regenerative capacity. Most cardiomyocytes live in a state of permanent cell-cycle arrest and are unable to re-enter the cycle. Cardiomyocytes switch from cell proliferation to a maturation state during neonatal development. Although several signaling pathways are involved in this transition, the molecular mechanisms by which these inputs coordinately regulate cardiomyocyte maturation are not fully understood. Retinoic acid (RA) plays a pivotal role in development, morphogenesis, and regeneration. Despite the importance of RA signaling in embryo heart development, little is known about its function in the early postnatal period. We found that mRNA expression of aldehyde dehydrogenase 1 family member A2 (Aldh1a2), which encodes the key enzyme for synthesizing all-trans retinoic acid (ATRA) and is an important regulator for RA signaling, was transiently upregulated in neonatal mouse ventricles. Single-cell transcriptome analysis and immunohistochemistry revealed that Aldh1a2 expression was enriched in cardiac fibroblasts during the early postnatal period. Administration of ATRA inhibited cardiomyocyte proliferation in cultured neonatal rat cardiomyocytes and human cardiomyocytes. RNA-seq analysis indicated that cell proliferation-related genes were downregulated in prenatal rat ventricular cardiomyocytes treated with ATRA, while cardiomyocyte maturation-related genes were upregulated. These findings suggest that RA signaling derived from cardiac fibroblasts is one of the key regulators of cardiomyocyte proliferation and maturation during neonatal heart development.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Cell Proliferation , Myocytes, Cardiac , Retinal Dehydrogenase , Signal Transduction , Tretinoin , Animals , Tretinoin/pharmacology , Tretinoin/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Mice , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Retinal Dehydrogenase/metabolism , Retinal Dehydrogenase/genetics , Cell Proliferation/drug effects , Rats , Humans , Up-Regulation , Animals, Newborn , Cell Cycle/drug effects , Cell Differentiation/drug effects , Heart/drug effects , Heart/growth & development , Cells, Cultured
10.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39082789

ABSTRACT

Skeletal muscles of the head and trunk originate in distinct lineages with divergent regulatory programmes converging on activation of myogenic determination factors. Branchiomeric head and neck muscles share a common origin with cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). The retinoic acid (RA) signalling pathway is required during a defined early time window for normal deployment of cells from posterior CPM to the heart. Here, we show that blocking RA signalling in the early mouse embryo also results in selective loss of the trapezius neck muscle, without affecting other skeletal muscles. RA signalling is required for robust expression of myogenic determination factors in posterior CPM and subsequent expansion of the trapezius primordium. Lineage-specific activation of a dominant-negative RA receptor reveals that trapezius development is not regulated by direct RA signalling to myogenic progenitor cells in CPM, or through neural crest cells, but indirectly through the somitic lineage, closely apposed with posterior CPM in the early embryo. These findings suggest that trapezius development is dependent on precise spatiotemporal interactions between cranial and somitic mesoderm at the head/trunk interface.


Subject(s)
Head , Mesoderm , Muscle Development , Neck Muscles , Signal Transduction , Tretinoin , Animals , Tretinoin/metabolism , Mice , Neck Muscles/embryology , Mesoderm/metabolism , Mesoderm/embryology , Head/embryology , Gene Expression Regulation, Developmental , Somites/metabolism , Somites/embryology , Receptors, Retinoic Acid/metabolism
11.
J Steroid Biochem Mol Biol ; 243: 106583, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38992392

ABSTRACT

The oviduct of the Chinese brown frog (Rana dybowskii) expands during pre-brumation rather than the breeding period, exhibiting a special physiological feature. Vitamin A is essential for the proper growth and development of many organisms, including the reproductive system such as ovary and oviduct. Vitamin A is metabolized into retinoic acid, which is crucial for oviduct formation. This study examined the relationship between oviducal expansion and vitamin A metabolism. We observed a significant increase in the weight and diameter of the oviduct in Rana dybowskii during pre-brumation. Vitamin A and its active metabolite, retinoic acid, notably increased during pre-brumation. The mRNA levels of retinol binding protein 4 (rbp4) and its receptor stra6 gene, involved in vitamin A transport, were elevated during pre-brumation compared to the breeding period. In the vitamin A metabolic pathway, the mRNA expression level of retinoic acid synthase aldh1a2 decreased significantly during pre-brumation, while the mRNA levels of retinoic acid α receptor (rarα) and the retinoic acid catabolic enzyme cyp26a1 increased significantly during pre-brumation, but not during the breeding period. Immunohistochemical results showed that Rbp4, Stra6, Aldh1a2, Rarα, and Cyp26a1 were expressed in ampulla region of the oviduct. Western blot results indicated that Aldh1a2 expression was lower, while Rbp4, Stra6, RARα, and Cyp26a1 were higher during pre-brumation compared to the breeding period. Transcriptome analyses further identified differential genes in the oviduct and found enrichment of differential genes in the vitamin A metabolism pathway, providing evidences for our study. These results suggest that the vitamin A metabolic pathway is more active during pre-brumation compared to the breeding period, and retinoic acid may regulate pre-brumation oviductal expansion through Rarα-mediated autocrine/paracrine modulation.


Subject(s)
Oviducts , Ranidae , Seasons , Vitamin A , Animals , Female , Vitamin A/metabolism , Oviducts/metabolism , Ranidae/metabolism , Ranidae/genetics , Tretinoin/metabolism
12.
Int J Mol Sci ; 25(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062755

ABSTRACT

Opsins are a class of transmembrane proteins encoded by opsin genes, and they play a variety of functional roles. Short wavelength-sensitive opsin 2 (sws2), one of the five classes of visual opsin genes, mainly senses blue light. Previous research has indicated that sws2 is essential for melanocyte formation in fish; however, its specific role in skin color differentiation remains to be elucidated. Here, we identified the sws2 gene in a prized reef-dwelling fish, Plectropomus leopardus. The full-length P. leopardus sws2 gene encodes a protein consisting of 351 amino acids, and exhibits substantial homology with other fish species. The expression of the sws2 gene was widespread across P. leopardus tissues, with high expression in eye and skin tissues. Through immunohistochemistry and in situ hybridization analyses, we discovered that the sws2 gene was primarily localized in the rod and cone cells of the retina, and epidermal cells of the skin. Furthermore, dsRNA interference was used for sws2 gene knockdown in living P. leopardus to elucidate its function in skin color differentiation. Black-color-related genes, melanin contents, and tyrosinase activity in the skin significantly decreased after sws2 knockdown (p < 0.05), but red-color-related genes and carotenoid and lutein contents significantly increased (p < 0.05). Retinoic acid injection produced the opposite results. Our results suggested that the sws2 gene influences P. leopardus skin color regulation by affecting vitamin synthesis and melanin-related gene expression levels. This study establishes a foundation for elucidating the molecular mechanisms by which sws2 regulates melanocyte formation in fish skin.


Subject(s)
Melanins , Skin , Tretinoin , Animals , Melanins/biosynthesis , Melanins/metabolism , Tretinoin/metabolism , Skin/metabolism , Fish Proteins/genetics , Fish Proteins/metabolism , Skin Pigmentation/genetics , Opsins/metabolism , Opsins/genetics , Gene Expression Regulation
13.
Toxicol Lett ; 398: 150-160, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38971454

ABSTRACT

Activation of pregnane X receptor (PXR) by xenobiotics has been associated with metabolic diseases. This study aimed to reveal the impact of PXR activation on hepatic metabolome and explore novel mechanisms underlying PXR-mediated lipid metabolism disorder in the liver. Wild-type and PXR-deficient male C57BL/6 mice were used as in vivo models, and hepatic steatosis was induced by pregnenolone-16α-carbonitrile, a typical rodent PXR agonist. Metabolomic analysis of liver tissues showed that PXR activation led to significant changes in metabolites involved in multiple metabolic pathways previously reported, including lipid metabolism, energy homeostasis, and amino acid metabolism. Moreover, the level of hepatic all-trans retinoic acid (ATRA), the main active metabolite of vitamin A, was significantly increased by PXR activation, and genes involved in ATRA metabolism exhibited differential expression following PXR activation or deficiency. Consistent with previous research, the expression of downstream target genes of peroxisome proliferator-activated receptor α (PPARα) was decreased. Analysis of fatty acids by Gas Chromatography-Mass Spectrometer further revealed changes in polyunsaturated fatty acid metabolism upon PXR activation, suggesting inhibition of PPARα activity. Taken together, our findings reveal a novel metabolomic signature of hepatic steatosis induced by PXR activation in mice.


Subject(s)
Fatty Acids, Unsaturated , Fatty Liver , Liver , Metabolomics , Mice, Inbred C57BL , PPAR alpha , Pregnane X Receptor , Tretinoin , Animals , Male , Pregnane X Receptor/metabolism , Pregnane X Receptor/genetics , Tretinoin/metabolism , Liver/metabolism , Liver/drug effects , Fatty Liver/metabolism , Fatty Liver/chemically induced , Fatty Acids, Unsaturated/metabolism , PPAR alpha/metabolism , PPAR alpha/genetics , Lipid Metabolism/drug effects , Mice , Mice, Knockout , Pregnenolone Carbonitrile/pharmacology , Disease Models, Animal
14.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000543

ABSTRACT

Human individual differences in brain cytochrome P450 (CYP) metabolism, including induction, inhibition, and genetic variation, may influence brain sensitivity to neurotoxins and thus participate in the onset of neurodegenerative diseases. The aim of this study was to explore the modulation of CYPs in neuronal cells. The experimental approach was focused on differentiating human neuroblastoma SH-SY5Y cells into a phenotype resembling mature dopamine neurons and investigating the effects of specific CYP isoform induction. The results demonstrated that the differentiation protocols using retinoic acid followed by phorbol esters or brain-derived neurotrophic factor successfully generated SH-SY5Y cells with morphological neuronal characteristics and increased neuronal markers (NeuN, synaptophysin, ß-tubulin III, and MAO-B). qRT-PCR and Western blot analysis showed that expression of the CYP 1A1, 3A4, 2D6, and 2E1 isoforms was detectable in undifferentiated cells, with subsequent increases in CYP 2E1, 2D6, and 1A1 following differentiation. Further increases in the 1A1, 2D6, and 2E1 isoforms following ß-naphthoflavone treatment and 1A1 and 2D6 isoforms following ethanol treatment were evident. These results demonstrate that CYP isoforms can be modulated in SH-SY5Y cells and suggest their potential as an experimental model to investigate the role of CYPs in neuronal processes involved in the development of neurodegenerative diseases.


Subject(s)
Cell Differentiation , Cytochrome P-450 Enzyme System , Neurodegenerative Diseases , Humans , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Cell Line, Tumor , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Tretinoin/pharmacology , Tretinoin/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroblastoma/genetics , Isoenzymes/metabolism , Isoenzymes/genetics , Dopaminergic Neurons/metabolism , Neurons/metabolism
15.
EMBO J ; 43(18): 3895-3915, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39060515

ABSTRACT

Dendritic cell (DC) dysfunction is known to exacerbate intestinal pathologies, but the mechanisms compromising DC-mediated immune regulation in this context remain unclear. Here, we show that intestinal dendritic cells from a mouse model of experimental colitis exhibit significant levels of noncanonical NF-κB signaling, which activates the RelB:p52 heterodimer. Genetic inactivation of this pathway in DCs alleviates intestinal pathologies in mice suffering from colitis. Deficiency of RelB:p52 diminishes transcription of Axin1, a critical component of the ß-catenin destruction complex, reinforcing ß-catenin-dependent expression of Raldh2, which imparts tolerogenic DC attributes by promoting retinoic acid synthesis. DC-specific impairment of noncanonical NF-κB signaling leads to increased colonic numbers of Tregs and IgA+ B cells, which promote luminal IgA production and foster eubiosis. Experimentally introduced ß-catenin haploinsufficiency in DCs with deficient noncanonical NF-κB signaling moderates Raldh2 activity, reinstating colitogenic sensitivity in mice. Finally, inflammatory bowel-disease patients also display a deleterious noncanonical NF-κB signaling signature in intestinal DCs. In sum, we establish how noncanonical NF-κB signaling in dendritic cells can subvert retinoic acid synthesis to fuel intestinal inflammation.


Subject(s)
Colitis , Dendritic Cells , NF-kappa B , Signal Transduction , beta Catenin , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , beta Catenin/metabolism , beta Catenin/genetics , NF-kappa B/metabolism , Colitis/immunology , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/genetics , Transcription Factor RelB/metabolism , Transcription Factor RelB/genetics , Retinal Dehydrogenase/metabolism , Retinal Dehydrogenase/genetics , Humans , Mice, Inbred C57BL , NF-kappa B p52 Subunit/metabolism , NF-kappa B p52 Subunit/genetics , Disease Models, Animal , Mice, Knockout , Immune Tolerance , Tretinoin/metabolism , Aldehyde Oxidoreductases
16.
Cells ; 13(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38994945

ABSTRACT

Spermatogenesis in mammalian testes is essential for male fertility, ensuring a continuous supply of mature sperm. The testicular microenvironment finely tunes this process, with retinoic acid, an active metabolite of vitamin A, serving a pivotal role. Retinoic acid is critical for various stages, including the differentiation of spermatogonia, meiosis in spermatogenic cells, and the production of mature spermatozoa. Vitamin A deficiency halts spermatogenesis, leading to the degeneration of numerous germ cells, a condition reversible with retinoic acid supplementation. Although retinoic acid can restore fertility in some males with reproductive disorders, it does not work universally. Furthermore, high doses may adversely affect reproduction. The inconsistent outcomes of retinoid treatments in addressing infertility are linked to the incomplete understanding of the molecular mechanisms through which retinoid signaling governs spermatogenesis. In addition to the treatment of male reproductive disorders, the role of retinoic acid in spermatogenesis also provides new ideas for the development of male non-hormone contraceptives. This paper will explore three facets: the synthesis and breakdown of retinoic acid in the testes, its role in spermatogenesis, and its application in male reproduction. Our discussion aims to provide a comprehensive reference for studying the regulatory effects of retinoic acid signaling on spermatogenesis and offer insights into its use in treating male reproductive issues.


Subject(s)
Spermatogenesis , Tretinoin , Male , Spermatogenesis/drug effects , Tretinoin/metabolism , Tretinoin/pharmacology , Humans , Animals , Reproduction/drug effects , Testis/metabolism , Testis/drug effects , Signal Transduction/drug effects , Infertility, Male/metabolism , Spermatozoa/metabolism , Spermatozoa/drug effects
17.
Biotechnol J ; 19(6): e2300659, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38863121

ABSTRACT

All-trans retinoic acid (atRA) is an endogenous ligand of the retinoic acid receptors, which heterodimerize with retinoid X receptors. AtRA is generated in tissues from vitamin A (retinol) metabolism to form a paracrine signal and is locally degraded by cytochrome P450 family 26 (CYP26) enzymes. The CYP26 family consists of three subtypes: A1, B1, and C1, which are differentially expressed during development. This study aims to develop and validate a high throughput screening assay to identify CYP26A1 inhibitors in a cell-free system using a luminescent P450-Glo assay technology. The assay performed well with a signal to background ratio of 25.7, a coefficient of variation of 8.9%, and a Z-factor of 0.7. To validate the assay, we tested a subset of 39 compounds that included known CYP26 inhibitors and retinoids, as well as positive and negative control compounds selected from the literature and/or the ToxCast/Tox21 portfolio. Known CYP26A1 inhibitors were confirmed, and predicted CYP26A1 inhibitors, such as chlorothalonil, prochloraz, and SSR126768, were identified, demonstrating the reliability and robustness of the assay. Given the general importance of atRA as a morphogenetic signal and the localized expression of Cyp26a1 in embryonic tissues, a validated CYP26A1 assay has important implications for evaluating the potential developmental toxicity of chemicals.


Subject(s)
High-Throughput Screening Assays , Retinoic Acid 4-Hydroxylase , High-Throughput Screening Assays/methods , Retinoic Acid 4-Hydroxylase/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Humans , Tretinoin/pharmacology , Tretinoin/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Reproducibility of Results
18.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891776

ABSTRACT

Neural tube defects (NTDs), which are caused by impaired embryonic neural tube closure, are one of the most serious and common birth defects. Peptidyl-prolyl cis/trans isomerase 1 (Pin1) is a prolyl isomerase that uniquely regulates cell signaling by manipulating protein conformation following phosphorylation, although its involvement in neuronal development remains unknown. In this study, we explored the involvement of Pin1 in NTDs and its potential mechanisms both in vitro and in vivo. The levels of Pin1 expression were reduced in NTD models induced by all-trans retinoic acid (Atra). Pin1 plays a significant role in regulating the apoptosis, proliferation, differentiation, and migration of neurons. Moreover, Pin1 knockdown significantly was found to exacerbate oxidative stress (OS) and endoplasmic reticulum stress (ERs) in neuronal cells. Further studies showed that the Notch1-Nrf2 signaling pathway may participate in Pin1 regulation of NTDs, as evidenced by the inhibition and overexpression of the Notch1-Nrf2 pathway. In addition, immunofluorescence (IF), co-immunoprecipitation (Co-IP), and GST pull-down experiments also showed that Pin1 interacts directly with Notch1 and Nrf2. Thus, our study suggested that the knocking down of Pin1 promotes NTD progression by inhibiting the activation of the Notch1-Nrf2 signaling pathway, and it is possible that this effect is achieved by disrupting the interaction of Pin1 with Notch1 and Nrf2, affecting their proteostasis. Our research identified that the regulation of Pin1 by retinoic acid (RA) and its involvement in the development of NTDs through the Notch1-Nrf2 axis could enhance our comprehension of the mechanism behind RA-induced brain abnormalities.


Subject(s)
NIMA-Interacting Peptidylprolyl Isomerase , Neural Tube Defects , Tretinoin , Animals , Female , Humans , Mice , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Neural Tube/metabolism , Neural Tube/drug effects , Neural Tube Defects/metabolism , Neural Tube Defects/genetics , Neural Tube Defects/chemically induced , Neurons/metabolism , Neurons/drug effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Oxidative Stress/drug effects , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Signal Transduction/drug effects , Tretinoin/metabolism , Tretinoin/pharmacology
19.
PLoS One ; 19(6): e0305350, 2024.
Article in English | MEDLINE | ID: mdl-38861553

ABSTRACT

All-trans retinoic acid (ATRA), recognized as the principal and most biologically potent metabolite of vitamin A, has been identified for its inhibitory effects on hepatitis B virus (HBV) replication. Nevertheless, the underlying mechanism remains elusive. The present study reveals that ATRA induces E6-associated protein (E6AP)-mediated proteasomal degradation of HBx to suppress HBV replication in human hepatoma cells in a p53-dependent pathway. For this effect, ATRA induced promoter hypomethylation of E6AP in the presence of HBx, which resulted in the upregulation of E6AP levels in HepG2 but not in Hep3B cells, emphasizing the p53-dependent nature of this effect. As a consequence, ATRA augmented the interaction between E6AP and HBx, resulting in substantial ubiquitination of HBx and consequent reduction in HBx protein levels in both the HBx overexpression system and the in vitro HBV replication model. Additionally, the knockdown of E6AP under ATRA treatment reduced the interaction between HBx and E6AP and decreased the ubiquitin-dependent proteasomal degradation of HBx, which prompted a recovery of HBV replication in the presence of ATRA, as confirmed by increased levels of intracellular HBV proteins and secreted HBV levels. This study not only contributes to the understanding of the complex interactions between ATRA, p53, E6AP, and HBx but also provides an academic basis for the clinical employment of ATRA in the treatment of HBV infection.


Subject(s)
Hepatitis B virus , Proteasome Endopeptidase Complex , Trans-Activators , Tretinoin , Tumor Suppressor Protein p53 , Ubiquitin-Protein Ligases , Viral Regulatory and Accessory Proteins , Virus Replication , Humans , Viral Regulatory and Accessory Proteins/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Proteasome Endopeptidase Complex/metabolism , Virus Replication/drug effects , Hepatitis B virus/physiology , Hepatitis B virus/drug effects , Tretinoin/pharmacology , Tretinoin/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Hep G2 Cells , Down-Regulation/drug effects , Ubiquitination/drug effects , Proteolysis/drug effects , Promoter Regions, Genetic , DNA Methylation/drug effects , Cell Line, Tumor
20.
Adv Exp Med Biol ; 1441: 683-696, 2024.
Article in English | MEDLINE | ID: mdl-38884742

ABSTRACT

During normal cardiovascular development, the outflow tract becomes septated and rotates so that the separate aorta and pulmonary trunk are correctly aligned with the left and right ventricles, respectively. However, when this process goes wrong, the aorta and pulmonary trunk are incorrectly positioned, resulting in oxygenated blood being directly returned to the lungs, with deoxygenated blood being delivered to the systemic circulation. This is termed transposition of the great arteries (TGA). The precise etiology of TGA is not known, but the use of animal models has elucidated that genes involved in determination of the left- embryonic body axis play key roles. Other factors such as retinoic acid levels are also crucial. This chapter reviews the animal models presenting with TGA that have been generated by genetic manipulation or with exogenous agents.


Subject(s)
Disease Models, Animal , Transposition of Great Vessels , Animals , Transposition of Great Vessels/genetics , Humans , Mice , Signal Transduction , Tretinoin/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL