Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Immunity ; 56(11): 2542-2554.e7, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37714152

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are crucial in promoting type 2 inflammation that contributes to both anti-parasite immunity and allergic diseases. However, the molecular checkpoints in ILC2s that determine whether to immediately launch a proinflammatory response are unknown. Here, we found that retinoid X receptor gamma (Rxrg) was highly expressed in small intestinal ILC2s and rapidly suppressed by alarmin cytokines. Genetic deletion of Rxrg did not impact ILC2 development but facilitated ILC2 responses and the tissue inflammation induced by alarmins. Mechanistically, RXRγ maintained the expression of its target genes that support intracellular cholesterol efflux, which in turn reduce ILC2 proliferation. Furthermore, RXRγ expression prevented ILC2 response to mild stimulations, including low doses of alarmin cytokine and mechanical skin injury. Together, we propose that RXRγ expression and its mediated lipid metabolic states function as a cell-intrinsic checkpoint that confers the threshold of ILC2 activation in the small intestine.


Subject(s)
Immunity, Innate , Retinoid X Receptor gamma , Humans , Alarmins , Lymphocytes , Inflammation , Cytokines/metabolism , Intestine, Small/metabolism
2.
Int J Mol Sci ; 23(10)2022 May 17.
Article in English | MEDLINE | ID: mdl-35628438

ABSTRACT

Zyxin is an LIM-domain-containing protein that regulates the assembly of F-actin filaments in cell contacts. Additionally, as a result of mechanical stress, Zyxin can enter nuclei and regulate gene expression. Previously, we found that Zyxin could affect mRNA stability of the maternally derived stemness factors of Pou5f3 family in Xenopus laevis embryos through binding to Y-box factor1. In the present work, we demonstrate that Zyxin can also affect mRNA stability of the maternally derived retinoid receptor Rxrγ through the same mechanism. Moreover, we confirmed the functional link between Zyxin and Rxrγ-dependent gene expression. As a result, Zyxin appears to play an essential role in the regulation of the retinoic acid signal pathway during early embryonic development. Besides, our research indicates that the mechanism based on the mRNA destabilization by Zyxin may take part in the control of the expression of a fairly wide range of maternal genes.


Subject(s)
RNA, Messenger, Stored , Tretinoin , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Retinoid X Receptor gamma , Signal Transduction , Tretinoin/pharmacology , Zyxin/genetics , Zyxin/metabolism
3.
Metab Brain Dis ; 37(5): 1351-1363, 2022 06.
Article in English | MEDLINE | ID: mdl-35486208

ABSTRACT

Cerebral ischemia is a common cerebrovascular disease with high mortality and disability rate. Exploring its mechanism is essential for developing effective treatment for cerebral ischemia. Therefore, this study aims to explore the regulatory effect and mechanism of retinoid X receptor γ (RXRγ) on cerebral ischemia-reperfusion (I/R) injury. A mouse intraluminal middle cerebral artery occlusion model was established, and PC12 cells were exposed to anaerobic/reoxygenation (A/R) as an in vitro model in this study. Cerebral I/R surgery or A/R treatment induced ferroptosis, downregulated RXRγ and GPX4 (glutathione peroxidase 4) levels, upregulated cyclooxygenase-2 (COX-2) level and increased ROS (reactive oxygen species) level in A/R induced cells or I/R brain tissues in vivo or PC12 cells in vitro. Knockdown of RXRγ downregulated GPX4 and increased COX-2 and ROS levels in A/R induced cells. RXRγ overexpression has the opposite effect. GPX4 knockdown reversed the improvement of RXRγ overexpression on COX-2 downregulation, GPX4 upregulation and ferroptosis in PC12 cells. Furthermore, chromatin immunoprecipitation (ChIP) and luciferase reporter gene assays revealed that RXRγ bound to GPX4 promoter region and activated its transcription. Overexpression of RXRγ or GPX4 alleviated brain damage and inhibited ferroptosis in I/R mice. In conclusion, RXRγ-mediated transcriptional activation of GPX4 might inhibit ferroptosis during I/R-induced brain injury.


Subject(s)
Brain Ischemia , Ferroptosis , Reperfusion Injury , Retinoid X Receptor gamma/metabolism , Animals , Brain Ischemia/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Mice , Neurons/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Rats , Reactive Oxygen Species/metabolism , Reperfusion , Reperfusion Injury/metabolism
4.
Biosci Rep ; 41(10)2021 10 29.
Article in English | MEDLINE | ID: mdl-34633445

ABSTRACT

To clarify the effect of retinoid X receptor-α/γ (RXR-α/γ) genes functional genetic variants (RXR-α rs4842194 G>A, RXR-γ rs100537 A>G and rs2134095 T>C) on the risk of gestational diabetes mellitus (GDM), a case-control study with 573 GDM patients and 740 pregnant women with normal glucose tolerance was performed in Guangxi area of China. An odds ratio (OR) with its corresponding 95% confidence interval (CI) was used to assess the strengths of the association between genetic variation and GDM. After adjustment of age and pre-BMI, the logistic regression analysis showed that the rs2134095 was significantly associated with GDM risk (CC vs. TT/TC: adjusted OR = 0.71, 95% CI = 0.56-0.90) in all subjects, and this result remained highly significant after Bonferroni's correction for multiple testing (P=0.004). The stratified analysis showed that rs2134095 was significantly associated with the risk of GDM among age > 30 years (adjusted OR = 0.61, 95% CI = 0.39-0.97), BMI > 22 kg/m2 (adjusted OR = 0.46, 95% CI = 0.30-0.70), systolic blood pressure (SBP) > 120 mmHg (adjusted OR = 1.96, 95% CI = 1.14-3.36), glycosylated hemoglobin A1c (HbA1c) < 6.5% (adjusted OR = 1.41, 95% CI = 1.11-1.78), TG ≤ 1.7 mmol/l (adjusted OR = 2.57, 95% CI = 1.45-4.53), TC ≤ 5.18 mmol/l (adjusted OR = 1.58, 95% CI = 1.13-2.22), high-density lipoprotein cholesterol (HDL-c) ≤ 1.5 mmol/l (adjusted OR = 1.70, 95% CI = 1.16-2.49) and low-density lipoprotein cholesterol (LDL-c) > 3.12 mmol/l (adjusted OR = 1.47, 95% CI = 1.08-2.00) subjects, under the recessive genetic model. We also found that rs2134095 interacted with age (Pinteraction=0.039), pre-BMI (Pinteraction=0.040) and TG (Pinteraction=0.025) influencing individual's genetic susceptibility to GDM. The rs2134095 T>C is significantly associated with the risk of GDM by effect of a single locus and/or complex joint gene-gene and gene-environment interactions. Larger sample-size and different population studies are required to confirm the findings.


Subject(s)
Diabetes, Gestational/genetics , Polymorphism, Single Nucleotide , Retinoid X Receptor alpha/genetics , Retinoid X Receptor gamma/genetics , Adult , Asian People/genetics , Biomarkers/blood , Blood Glucose/genetics , Blood Glucose/metabolism , Case-Control Studies , China/epidemiology , Diabetes, Gestational/blood , Diabetes, Gestational/diagnosis , Diabetes, Gestational/ethnology , Female , Genetic Association Studies , Genetic Predisposition to Disease , Glycated Hemoglobin/metabolism , Humans , Lipids/blood , Phenotype , Pregnancy , Risk Assessment , Risk Factors
5.
Nature ; 598(7881): 483-488, 2021 10.
Article in English | MEDLINE | ID: mdl-34599305

ABSTRACT

The prefrontal cortex (PFC) and its connections with the mediodorsal thalamus are crucial for cognitive flexibility and working memory1 and are thought to be altered in disorders such as autism2,3 and schizophrenia4,5. Although developmental mechanisms that govern the regional patterning of the cerebral cortex have been characterized in rodents6-9, the mechanisms that underlie the development of PFC-mediodorsal thalamus connectivity and the lateral expansion of the PFC with a distinct granular layer 4 in primates10,11 remain unknown. Here we report an anterior (frontal) to posterior (temporal), PFC-enriched gradient of retinoic acid, a signalling molecule that regulates neural development and function12-15, and we identify genes that are regulated by retinoic acid in the neocortex of humans and macaques at the early and middle stages of fetal development. We observed several potential sources of retinoic acid, including the expression and cortical expansion of retinoic-acid-synthesizing enzymes specifically in primates as compared to mice. Furthermore, retinoic acid signalling is largely confined to the prospective PFC by CYP26B1, a retinoic-acid-catabolizing enzyme, which is upregulated in the prospective motor cortex. Genetic deletions in mice revealed that retinoic acid signalling through the retinoic acid receptors RXRG and RARB, as well as CYP26B1-dependent catabolism, are involved in proper molecular patterning of prefrontal and motor areas, development of PFC-mediodorsal thalamus connectivity, intra-PFC dendritic spinogenesis and expression of the layer 4 marker RORB. Together, these findings show that retinoic acid signalling has a critical role in the development of the PFC and, potentially, in its evolutionary expansion.


Subject(s)
Organogenesis , Prefrontal Cortex/embryology , Prefrontal Cortex/metabolism , Tretinoin/metabolism , Animals , Axons/metabolism , Cerebral Cortex , Down-Regulation , Female , Humans , Macaca mulatta , Male , Mice , Pan troglodytes , Prefrontal Cortex/anatomy & histology , Prefrontal Cortex/cytology , Receptors, Retinoic Acid/deficiency , Retinoid X Receptor gamma/deficiency , Signal Transduction , Synapses/metabolism , Thalamus/anatomy & histology , Thalamus/cytology , Thalamus/metabolism
6.
Sci Rep ; 11(1): 5552, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692389

ABSTRACT

Retinoid X receptors are members of the nuclear receptor family that regulate gene expression in response to retinoic acid and related ligands. Group 1 metabotropic glutamate receptors are G-protein coupled transmembrane receptors that activate intracellular signaling cascades in response to the neurotransmitter, glutamate. These two classes of molecules have been studied independently and found to play important roles in regulating neuronal physiology with potential clinical implications for disorders such as depression, schizophrenia, Parkinson's and Alzheimer's disease. Here we show that mice lacking the retinoid X receptor subunit, RXRγ, exhibit impairments in group 1 mGluR-mediated electrophysiological responses at hippocampal Schaffer collateral-CA1 pyramidal cell synapses, including impaired group 1 mGluR-dependent long-term synaptic depression (LTD), reduced group 1 mGluR-induced calcium release, and loss of group 1 mGluR-activated voltage-sensitive currents. These animals also exhibit impairments in a subset of group 1 mGluR-dependent behaviors, including motor performance, spatial object recognition, and prepulse inhibition. Together, these observations demonstrate convergence between the RXRγ and group 1 mGluR signaling pathways that may function to coordinate their regulation of neuronal activity. They also identify RXRγ as a potential target for the treatment of disorders in which group 1 mGluR signaling has been implicated.


Subject(s)
CA1 Region, Hippocampal/metabolism , Long-Term Synaptic Depression , Pyramidal Cells/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinoid X Receptor gamma/metabolism , Signal Transduction , Synapses/metabolism , Animals , Mice , Mice, Knockout , Receptors, Metabotropic Glutamate/genetics , Retinoid X Receptor gamma/genetics , Synapses/genetics
7.
Stem Cells ; 39(4): 414-428, 2021 04.
Article in English | MEDLINE | ID: mdl-33400844

ABSTRACT

Organoid cultures represent a unique tool to investigate the developmental complexity of tissues like the human retina. NRL is a transcription factor required for the specification and homeostasis of mammalian rod photoreceptors. In Nrl-deficient mice, photoreceptor precursor cells do not differentiate into rods, and instead follow a default photoreceptor specification pathway to generate S-cone-like cells. To investigate whether this genetic switch mechanism is conserved in humans, we used CRISPR/Cas9 gene editing to engineer an NRL-deficient embryonic stem cell (ESC) line (NRL-/- ), and differentiated it into retinal organoids. Retinal organoids self-organize and resemble embryonic optic vesicles (OVs) that recapitulate the natural histogenesis of rods and cone photoreceptors. NRL-/- OVs develop comparably to controls, and exhibit a laminated, organized retinal structure with markers of photoreceptor synaptogenesis. Using immunohistochemistry and quantitative polymerase chain reaction (qPCR), we observed that NRL-/- OVs do not express NRL, or other rod photoreceptor markers directly or indirectly regulated by NRL. On the contrary, they show an abnormal number of photoreceptors positive for S-OPSIN, which define a primordial subtype of cone, and overexpress other cone genes indicating a conserved molecular switch in mammals. This study represents the first evidence in a human in vitro ESC-derived organoid system that NRL is required to define rod identity, and that in its absence S-cone-like cells develop as the default photoreceptor cell type. It shows how gene edited retinal organoids provide a useful system to investigate human photoreceptor specification, relevant for efforts to generate cells for transplantation in retinal degenerative diseases.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Eye Proteins/genetics , Human Embryonic Stem Cells/metabolism , Organoids/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Base Sequence , Basic-Leucine Zipper Transcription Factors/deficiency , CRISPR-Cas Systems , Cell Differentiation , Exons , Gene Editing/methods , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Human Embryonic Stem Cells/cytology , Humans , Opsins/genetics , Opsins/metabolism , Organoids/pathology , Recoverin/genetics , Recoverin/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinoid X Receptor gamma/genetics , Retinoid X Receptor gamma/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
8.
Curr Mol Med ; 20(6): 408-414, 2020.
Article in English | MEDLINE | ID: mdl-31808386

ABSTRACT

OBJECTIVE: To investigate the relationship between genetic polymorphisms of RXRG rs1467664, rs3753898 and the genetic susceptibility of type 2 diabetes in the Chinese Han population from South China. METHODS: In our case-control study, single-nucleotide polymorphisms (SNPs) rs1467664 and rs3753898 were genotyped by SNPscanTM kit in 1092 patients with T2D as cases and 1092 normal persons as controls. The distributions of genotype and allele frequencies in two groups were analyzed by the SPSS 20.0 software. RESULTS: The distribution of genotypes and alleles of RXRG rs3753898 was statistically significant between the two groups, but there was no significant difference in the distribution of genotypes and alleles of the rs1467664. Before and after the adjustment of age, sex and BMI, rs3753898 in the two groups had statistical significance under the additive, dominant and recessive models (P<0.05), but no statistical differences were found under the overdominance and co-dominant genetic models (P>0.05). There was no significant difference in the genetic models of rs1467664 between the two groups (P>0.05). The haplotype, which consists of rs1467664 allele T and rs3753898 allele A was a high-risk factor for T2D, OR=1.27, 95% CI (1.09-1.47), Padj=0.002. CONCLUSION: Our results showed that the single nucleotide polymorphism of RXRG rs3753898 may be related to genetic susceptibility of type 2 diabetes. The haplotype consisting of the allele T of rs1467664 and the allele A of rs3753898 is a risk factor for type 2 diabetes, suggesting that the genetic variation of RXRG gene may be the genetic cause of diabetes mellitus in the Chinese Han population.


Subject(s)
Polymorphism, Single Nucleotide/genetics , Retinoid X Receptor gamma/genetics , Aged , Alleles , Asian People , Case-Control Studies , Diabetes Mellitus, Type 2/genetics , Female , Gene Frequency/genetics , Genetic Association Studies , Genetic Predisposition to Disease/genetics , Genotype , Haplotypes/genetics , Humans , Linkage Disequilibrium/genetics , Male , Middle Aged , Software
9.
J Steroid Biochem Mol Biol ; 198: 105571, 2020 04.
Article in English | MEDLINE | ID: mdl-31881311

ABSTRACT

The retinoid X receptor (RXR) is a member of the nuclear receptor (NR) superfamily that occupies the central position among other NRs by forming both homodimers and heterodimers with other representatives of the family. RXR shares similar structural domains with other members of NRs. The major differences in the subtypes and isoforms of RXR are in the AB region. To date, there have been no data concerning the molecular properties of the AB region of hRXRγ (AB_hRXG). Here, we describe the biochemical and biophysical properties of the recombinant AB_hRXG. The results indicate that AB_hRXG shows the structural and functional characteristics of the pre-molten globule-like (PMG-like) group of intrinsically disordered proteins (IDPs) and also has a significant propensity for folding. We also present the first experimental evidence showing that the AB region of NRs promotes the formation of liquid-liquid phase separation (LLPS).


Subject(s)
Intrinsically Disordered Proteins/chemistry , Retinoid X Receptor gamma/chemistry , Circular Dichroism , Computer Simulation , Humans , Liquid-Liquid Extraction , Protein Binding , Protein Denaturation , Protein Folding , Protein Multimerization , Protein Structure, Secondary , Temperature
10.
Cells ; 8(12)2019 12 04.
Article in English | MEDLINE | ID: mdl-31817149

ABSTRACT

Skeletal muscle, the largest part of the total body mass, influences energy and protein metabolism as well as maintaining homeostasis. Herein, we demonstrate that during murine muscle satellite cell and myoblast differentiation, transthyretin (TTR) can exocytose via exosomes and enter cells as TTR- thyroxine (T4) complex, which consecutively induces the intracellular triiodothyronine (T3) level, followed by T3 secretion out of the cell through the exosomes. The decrease in T3 with the TTR level in 26-week-old mouse muscle, compared to that in 16-week-old muscle, suggests an association of TTR with old muscle. Subsequent studies, including microarray analysis, demonstrated that T3-regulated genes, such as FNDC5 (Fibronectin type III domain containing 5, irisin) and RXRγ (Retinoid X receptor gamma), are influenced by TTR knockdown, implying that thyroid hormones and TTR coordinate with each other with respect to muscle growth and development. These results suggest that, in addition to utilizing T4, skeletal muscle also distributes generated T3 to other tissues and has a vital role in sensing the intracellular T4 level. Furthermore, the results of TTR function with T4 in differentiation will be highly useful in the strategic development of novel therapeutics related to muscle homeostasis and regeneration.


Subject(s)
Cell Differentiation , Muscle Development , Myoblasts/metabolism , Prealbumin/metabolism , Thyroid Hormones/metabolism , Animals , Cell Line , Cells, Cultured , Fibronectins/genetics , Fibronectins/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myoblasts/cytology , Prealbumin/genetics , Retinoid X Receptor gamma/genetics , Retinoid X Receptor gamma/metabolism
11.
Br J Cancer ; 121(9): 776-785, 2019 10.
Article in English | MEDLINE | ID: mdl-31558802

ABSTRACT

BACKGROUND: Retinoid X Receptor Gamma (RXRG) is a member of the nuclear receptor superfamily and plays a role in tumour suppression. This study aims to explore the prognostic significance of RXRG in breast cancer. METHODS: Primary breast cancer tissue microarrays (n = 923) were immuno-stained for RXRG protein and correlated with clinicopathological features, and patient outcome. RESULTS: Nuclear RXRG expression was significantly associated with smaller tumour size (p = 0.036), lower grade (p < 0.001), lobular histology (p = 0.016), lower Nottingham Prognostic Index (p = 0.04) and longer breast cancer-specific survival (p < 0.001), and longer time to distant metastasis (p = 0.002). RXRG expression showed positive association with oestrogen receptor (ER)-related biomarkers: GATA3, FOXA1, STAT3 and MED7 (all p < 0.001) and a negative correlation with the Ki67 proliferation marker. Multivariate analysis demonstrated RXRG protein as an independent predictor of longer breast cancer-specific survival and distant metastasis-free survival. In the external validation cohorts, RXRG expression was associated with improved patients' outcome (p = 0.025). In ER-positive tumours, high expression of RXRG was associated with better patient outcome regardless of adjuvant systemic therapy. ER signalling pathway was the top predicted master regulator of RXRG protein expression (p = 0.005). CONCLUSION: This study provides evidence for the prognostic value of RXRG in breast cancer particularly the ER-positive tumours.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Estrogen/metabolism , Retinoid X Receptor gamma/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cohort Studies , Female , Gene Expression , Humans , Immunohistochemistry , Neoplasm Invasiveness , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinoid X Receptor gamma/biosynthesis , Retinoid X Receptor gamma/genetics , Tissue Array Analysis
12.
Cells ; 8(8)2019 08 06.
Article in English | MEDLINE | ID: mdl-31390799

ABSTRACT

Remyelination in the adult brain relies on the reactivation of the Neuronal Precursor Cell (NPC) niche and differentiation into Oligodendrocyte Precursor Cells (OPCs) as well as on OPC maturation into myelinating oligodendrocytes (OLs). These two distinct phases in OL development are defined by transcriptional and morphological changes. How this differentiation program is controlled remains unclear. We used two drugs that stimulate myelin basic protein (MBP) expression (Clobetasol and Gefitinib) alone or combined with epidermal growth factor receptor (EGFR) or Retinoid X Receptor gamma (RXRγ) gene silencing to decode the receptor signaling required for OPC differentiation in myelinating OLs. Electrospun polystyrene (PS) microfibers were used as synthetic axons to study drug efficacy on fiber engagement. We show that EGFR inhibition per se stimulates MBP expression and increases Clobetasol efficacy in OPC differentiation. Consistent with this, Clobetasol and Gefitinib co-treatment, by co-regulating RXRγ, MBP and phosphatidylinositol 4,5-bisphosphate (PIP2) levels, maximizes synthetic axon engagement. Conversely, RXRγ gene silencing reduces the ability of the drugs to promote MBP expression. This work provides a view of how EGFR/ErbB inhibition controls OPC differentiation and indicates the combination of Clobetasol and Gefitinib as a potent remyelination-enhancing treatment.


Subject(s)
Clobetasol/pharmacology , ErbB Receptors/metabolism , Gefitinib/pharmacology , Myelin Basic Protein/metabolism , Oligodendrocyte Precursor Cells , Oligodendroglia , Retinoid X Receptor gamma/metabolism , Animals , Cell Differentiation , Cell Line , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Remyelination
13.
Methods Mol Biol ; 2019: 1-14, 2019.
Article in English | MEDLINE | ID: mdl-31359385

ABSTRACT

Retinoid and rexinoid receptors are known to regulate key processes during development, differentiation, and cell death in vertebrates. However, their contributions to progression of malignant disease remain largely elusive although it is realized that transformed cancer cells, which essentially evade apoptosis, may display altered molecular expressions or functions associated with retinoid signaling. Here, using a progression model of ovarian cancer, we describe a proteomics-based approach including experimental procedures toward identification and validation of altered protein profiles during transformation. Effectively, this specifies loss of RXR-γ during progression of epithelial ovarian cancer.


Subject(s)
Carcinoma, Ovarian Epithelial/pathology , Ovarian Neoplasms/pathology , Proteomics/methods , Retinoid X Receptor gamma/deficiency , Animals , Carcinoma, Ovarian Epithelial/metabolism , Cell Line, Tumor , Disease Progression , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Stem Cell Res ; 37: 101443, 2019 05.
Article in English | MEDLINE | ID: mdl-31022610

ABSTRACT

Oligodendrocyte precursor cells (OPCs) differentiation from multipotent neural stem cells (NSCs) into mature oligodendrocytes is driven by thyroid hormone and mediated by thyroid hormone receptors (TRs). We show that several nuclear receptors display strong changes in expression levels between fetal and adult NSCs, with an overexpression of TRß and a lower expression of RXRγ in adult. Such changes may determine the reduced capacity of adult OPCs to differentiate as supported by reduced yield of maturation and compromised mRNA expression of key genes. RXRγ may be the determinant of these differences, on the evidence of reduced number of mature oligodendrocytes and increased number of proliferating OPCs in RXRγ-/- cultures. Such data also points to RXRγ as an important regulator of the cell cycle exit, as proved by the dysregulation of T3-induced cell cycle exit-related genes. Our data highlight the biological differences between fetal and adult OPCs and demonstrate the essential role of RXRγ in the T3-mediated OPCs maturation process.


Subject(s)
Adult Stem Cells/cytology , Cell Differentiation , Fetus/cytology , Neural Stem Cells/cytology , Neurogenesis , Oligodendrocyte Precursor Cells/cytology , Retinoid X Receptor gamma/physiology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Cells, Cultured , Fetus/drug effects , Fetus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Thyroid Hormones/pharmacology
15.
Cell ; 174(4): 843-855.e19, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30017245

ABSTRACT

Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Melanoma/drug therapy , Neoplasm, Residual/drug therapy , Neoplastic Stem Cells/drug effects , Neural Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Retinoid X Receptor gamma/antagonists & inhibitors , Animals , Biomarkers, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Male , Melanoma/metabolism , Melanoma/pathology , Mice, SCID , Mutation , Neoplasm, Residual/metabolism , Neoplasm, Residual/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Int J Mol Med ; 42(4): 2042-2052, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30015907

ABSTRACT

The elucidation of the underlying molecular mechanism of H2O2­induced adipocyte differentiation in mesenchymal stem cells (MSCs) is important for the development of treatments for metabolic diseases. The aim of the present study was to identify microRNA (miR)­330­5p, which targets retinoid X receptor γ (RXRγ) and to determine the function of H2O2­induced adipogenic differentiation of MSCs. During differentiation of MSCs into adipocytes induced by H2O2, miR­330­5p expression was decreased with a concomitant increase in RXRγ expression. A luciferase assay with RXRγ 3'­untranslated region (UTR) reporter plasmid, including the miR­330­5p­binding sequences, identified that the introduction of miR­330­5p decreases luciferase activity. However, it did not affect the activity of mutated RXRγ 3'­UTR reporter. Enforced expression of miR­330­5p significantly inhibited adipocyte differentiation by decreasing RXRγ mRNA and protein levels. In contrast, inhibition of the endogenous miR­330­5p promoted the formation of lipid droplets by rescuing RXRγ expression. Furthermore, the effects of inhibition of RXRγ were similar to those of overexpression of miR­330­5p on H2O2­induced adipogenic differentiation from MSCs. miR­330­5p inhibits H2O2­induced adipogenic differentiation of MSCs, and this is dependent on RXRγ. Taken together, the results of the present study revealed that miR­330­5p acts as a critical regulator of RXRγ, and is able to determinate the fate of MSCs to differentiate into adipocytes. This suggests that miR­330­5p and RXRγ may be target molecules for controlling metabolic diseases.


Subject(s)
Adipogenesis/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Retinoid X Receptor gamma/biosynthesis , Animals , Cells, Cultured , Rats , Rats, Sprague-Dawley
17.
Epigenetics ; 13(3): 264-274, 2018.
Article in English | MEDLINE | ID: mdl-29465290

ABSTRACT

Gene regulatory analysis of highly diverse human tissues in vivo is essentially constrained by the challenge of performing genome-wide, integrated epigenetic and transcriptomic analysis in small selected groups of specific cell types. Here we performed genome-wide bisulfite sequencing and RNA-seq from the same small groups of bronchial and alveolar cells isolated by laser capture microdissection from flash-frozen lung tissue of 12 donors and their peripheral blood T cells. Methylation and transcriptome patterns differed between alveolar and bronchial cells, while each of these epithelia showed more differences from mesodermally-derived T cells. Differentially methylated regions (DMRs) between alveolar and bronchial cells tended to locate at regulatory regions affecting promoters of 4,350 genes. A large number of pathways enriched for these DMRs including GTPase signal transduction, cell death, and skeletal muscle. Similar patterns of transcriptome differences were observed: 4,108 differentially expressed genes (DEGs) enriched in GTPase signal transduction, inflammation, cilium assembly, and others. Prioritizing using DMR-DEG regulatory network, we highlighted genes, e.g., ETS1, PPARG, and RXRG, at prominent alveolar vs. bronchial cell discriminant nodes. Our results show that multi-omic analysis of small, highly specific cells is feasible and yields unique physiologic loci distinguishing human lung cell types in situ.


Subject(s)
DNA Methylation/genetics , Lung/metabolism , PPAR gamma/genetics , Proto-Oncogene Protein c-ets-1/genetics , Retinoid X Receptor gamma/genetics , Alveolar Epithelial Cells/metabolism , Cell Lineage/genetics , Epigenesis, Genetic , GTP Phosphohydrolases/genetics , Gene Regulatory Networks/genetics , Genome, Human/genetics , Humans , Laser Capture Microdissection , Lung/cytology , Promoter Regions, Genetic , Signal Transduction , T-Lymphocytes/metabolism , Transcriptome/genetics , Whole Genome Sequencing
18.
J Anim Physiol Anim Nutr (Berl) ; 102(1): e279-e287, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28503816

ABSTRACT

This study was designed to investigate effects of xanthophylls on serum lipid profile (triglyceride, TG; cholesterol, CHO; high-density lipoprotein cholesterol, HDLC; and low-density lipoprotein cholesterol, LDLC) and nuclear factor (peroxisome proliferator-activated receptor gamma, PPARγ; PPAR gamma coactivator 1 alpha, PGC1α; retinoid X receptor gamma, RXRγ; and retinoic acid receptor alpha, RARα) gene expression of breeding hens and chicks. In experiment 1, 432 hens were divided into three groups and fed diets supplemented with 0 (as control group), 20 or 40 mg/kg xanthophylls. Blood was sampled at 7, 14, 21, 28 and 35 days of trial. Liver, duodenum, jejunum and ileum were sampled at 35 days of trial. Results showed that serum HDLC level of hens was increased after dietary 40 mg/kg xanthophyll addition for 21, 28 and 35 days, while serum TG, CHO and LDLC were not affected. Xanthophyll addition also increased PPARγ expression in jejunum, RXRγ expression in duodenum and jejunum, and RARα expression in liver and duodenum. Experiment 2 was a 2 × 2 factorial design. Male chicks hatched from 0 or 40 mg/kg xanthophyll diet of hens were fed diet containing either 0 or 40 mg/kg xanthophylls. Liver, duodenum, jejunum and ileum were sampled at 0, 7, 14 and 21 days after hatching. Blood samples were also collected at 21 days. Results showed that in ovo xanthophylls elevated PPARγ in duodenum and jejunum, and RXRγ and RARα in liver of chicks mainly within 1 week after hatching, while dietary xanthophylls increased serum HDLC level and PPARγ and RXRγ in liver from 2 weeks onwards. In conclusion, our research suggested xanthophylls can regulate serum lipid profile and nuclear factor expression in hens and chicks.


Subject(s)
Chickens/metabolism , Cholesterol, HDL/blood , PPAR gamma/metabolism , Retinoid X Receptor gamma/metabolism , Xanthophylls/pharmacology , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Chickens/blood , Diet/veterinary , Dietary Supplements , Female , Gene Expression Regulation/drug effects , Male , PPAR gamma/genetics , Retinoid X Receptor alpha , Retinoid X Receptor gamma/genetics
19.
Mol Ther ; 25(3): 634-653, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28143742

ABSTRACT

The cone function is essential to mediate high visual acuity, color vision, and daylight vision. Inherited cone dystrophies and age-related macular degeneration affect a substantial percentage of the world population. To identify and isolate the most competent cells for transplantation and integration into the retina, cone tracing during development would be an important added value. To that aim, the Chrnb4-EGFP mouse line was characterized throughout retinogenesis. It revealed a sub-population of early retinal progenitors expressing the reporter gene that is progressively restricted to mature cones during retina development. The presence of the native CHRNB4 protein was confirmed in EGFP-positive cells, and it presents a similar pattern in the human retina. Sub-retinal transplantations of distinct subpopulations of Chrnb4-EGFP-expressing cells revealed the embryonic day 15.5 high-EGFP population the most efficient cells to interact with host retinas to provoke the appearance of EGFP-positive cones in the photoreceptor layer. Importantly, transplantations into the DsRed retinas revealed material exchanges between donor and host retinas, as >80% of transplanted EGFP-positive cones also were DsRed positive. Whether this cell material fusion is of significant therapeutic advantage requires further thorough investigations. The Chrnb4-EGFP mouse line definitely opens new research perspectives in cone genesis and retina repair.


Subject(s)
Cell Tracking/methods , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Nicotinic/genetics , Recombinant Fusion Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Animals , Humans , Macular Degeneration , Mice , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Retina/embryology , Retina/metabolism , Retinoid X Receptor gamma/genetics , Retinoid X Receptor gamma/metabolism , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism
20.
Cell Rep ; 18(3): 624-635, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099842

ABSTRACT

Brown adipose tissue (BAT) has attracted considerable research interest because of its therapeutic potential to treat obesity and associated metabolic diseases. Augmentation of brown fat mass and/or its function may represent an attractive strategy to enhance energy expenditure. Using high-throughput phenotypic screening to induce brown adipocyte reprogramming in committed myoblasts, we identified a retinoid X receptor (RXR) agonist, bexarotene (Bex), that efficiently converted myoblasts into brown adipocyte-like cells. Bex-treated mice exhibited enlarged BAT mass, enhanced BAT function, and a modest browning effect in subcutaneous white adipose tissue (WAT). Expression analysis showed that Bex initiated several "browning" pathways at an early stage during brown adipocyte reprogramming. Our findings suggest RXRs as new master regulators that control brown and beige fat development and activation, unlike the common adipogenic regulator PPARγ. Moreover, we demonstrated that selective RXR activation may potentially offer a therapeutic approach to manipulate brown/beige fat function in vivo.


Subject(s)
Adipose Tissue, Brown/metabolism , Cellular Reprogramming/genetics , Adipogenesis/drug effects , Adipose Tissue, Brown/cytology , Adipose Tissue, White/metabolism , Animals , Bexarotene , Body Weight/drug effects , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Oxygen Consumption/drug effects , PPAR gamma/metabolism , RNA Interference , Retinoid X Receptor alpha/antagonists & inhibitors , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Retinoid X Receptor beta/antagonists & inhibitors , Retinoid X Receptor beta/genetics , Retinoid X Receptor beta/metabolism , Retinoid X Receptor gamma/antagonists & inhibitors , Retinoid X Receptor gamma/genetics , Retinoid X Receptor gamma/metabolism , Tetrahydronaphthalenes/pharmacology , Thermogenesis/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL