Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
J Exp Clin Cancer Res ; 40(1): 141, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33902658

ABSTRACT

BACKGROUND: Retinoids are promising agents in the treatment of different types of neoplasia including estrogen receptor-positive breast cancers, whereas refractoriness/low sensitivity is observed in triple-negative breast cancer (TNBC) subtype. However, the reason for these diverse retinoid-sensitivity remains elusive. METHODS: Determinants of retinoid sensitivity were investigated using immunohistochemistry of primary patient samples, and identified retinoic acid receptor α (RARα) as a putative factor. The anti-tumor activity of hypo-phosphorylated RARα was investigated in TNBC cell models and a xenograft mouse model. Next, miRNA sequencing analysis was performed to identify the target miRNA of RARα, and luciferase reporter was used to confirm the direct target gene of miR-3074-5p. RESULTS: We discovered that serine-77 residue of RARα was constantly phosphorylated, which correlated with TNBC's resistance to retinoids. Overexpression of a phosphorylation-defective mutant RARαS77A mimicked activated RARα and repressed TNBC cell progression both in vitro and in vivo, via activating cell cycle arrest, apoptosis, and cytotoxic autophagy, independent of RARα agonists. We further revealed that the anti-tumor action of RARαS77A was, at least in part, mediated by the up-regulation of miR-3074-5p, which directly targeted DHRS3, a reductase negatively associated with TNBC patient survival. Our results suggest that the inhibition of RARαS77 phosphorylation by either expressing RARαS77A or inhibiting RARα's phosphokinase CDK7, can bypass RA stimuli to transactivate tumor-suppressive miR-3074-5p and reduce oncogenic DHRS3, thus overcoming the RA-resistance of TNBC. CONCLUSION: The novel regulatory network, involving RARαS77 phosphorylation, miR-3074-5p, and DHRS3, emerges as a new target for TNBC treatment.


Subject(s)
Alcohol Oxidoreductases/metabolism , MicroRNAs/metabolism , Retinoic Acid Receptor alpha/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Growth Processes/physiology , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm , Female , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Retinoic Acid Receptor alpha/metabolism , Tretinoin/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology
2.
Dev Dyn ; 250(7): 1036-1050, 2021 07.
Article in English | MEDLINE | ID: mdl-33452727

ABSTRACT

BACKGROUND: Pharyngeal arches (PA) are sequentially generated in an anterior-to-posterior order. Ripply3 is essential for posterior PA development in mouse embryos and its expression is sequentially activated in ectoderm and endoderm prior to formation of each PA. Since the PA phenotype of Ripply3 knockout (KO) mice is similar to that of retinoic acid (RA) signal-deficient embryos, we investigated the relationship between RA signaling and Ripply3 in mouse embryos. RESULTS: In BMS493 (pan-RAR antagonist) treated embryos, which are defective in third and fourth PA development, Ripply3 expression is decreased in the region posterior to PA2 at E9.0. This expression remains and its distribution is expanded posteriorly at E9.5. Conversely, high dose RA exposure does not apparently change its expression at E9.0 and 9.5. Knockout of retinaldehyde dehydrogenase 2 (Raldh2), which causes more severe PA defect, attenuates sequential Ripply3 expression at PA1 and reduces its expression level. EGFP reporter expression driven by a 6 kb Ripply3 promoter fragment recapitulates the endogenous Ripply3 mRNA expression during PA development in wild-type, but its distribution is expanded posteriorly in BMS493-treated and Raldh2 KO embryos. CONCLUSION: Spatio-temporal regulation of Ripply3 expression by RA signaling is indispensable for the posterior PA development in mouse.


Subject(s)
Branchial Region/embryology , Repressor Proteins/genetics , Tretinoin/metabolism , Animals , Benzoates/pharmacology , Branchial Region/drug effects , Branchial Region/metabolism , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/drug effects , Mice , Mice, Inbred ICR , Mice, Transgenic , Morphogenesis/drug effects , Morphogenesis/genetics , Pregnancy , Repressor Proteins/metabolism , Retinoic Acid Receptor alpha/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Stilbenes/pharmacology , Tretinoin/pharmacology , Tretinoin/physiology
3.
Drug Des Devel Ther ; 14: 297-308, 2020.
Article in English | MEDLINE | ID: mdl-32158187

ABSTRACT

BACKGROUND: Hypervitaminosis A, alcoholism or medical treatment for acute promyelocytic leukaemia may cause unphysiologically high accumulation of all-trans retinoic acid (ATRA), which could inhibit osteoblastogenesis, thereby triggering osteoporosis. We have shown that bone morphogenetic protein-2 (BMP-2) can only partially antagonize the inhibitive effects of ATRA. In this study, we hypothesized that antagonists of retinoic acid receptors (RARs) could further antagonize the inhibitive effect of ATRA and rescue BMP2-induced osteoblastogenesis. MATERIALS AND METHODS: We first screened the dose-dependent effects of the specific antagonists of RAR α, ß and γ and transforming growth factor-beta receptor (ER-50891, LE-135, MM11253, and SB-43142, respectively) on ATRA-induced inhibition of the total cell metabolic activity and proliferation of preosteoblasts. We selected ER-50891 and tested its effects on osteoblastogenesis with the presence or absence of 1 µM ATRA and/or 200 ng/mL BMP-2. We measured the following parameters: Alkaline phosphatase activity (ALP), osteocalcin (OCN) expression and extracellular matrix mineralization as well as the level of phosphorylated Smad1/5. RESULTS: ER-50891 but not LE-135, MM11253, or SB-431542 significantly antagonized the inhibition of ATRA and enhanced the total cell metabolic activity and proliferation of preosteoblasts. Dose-dependent assays show ER-50891 could also rescue ATRA inhibited OCN expression and mineralization with or without the induction of BMP. ER-50891 also suppressed the ALP activity that was synergistically enhanced by BMP and ATRA. Neither ATRA, nor ER-50891 or their combination significantly affected the level of BMP-induced phosphorylated Smad1/5. CONCLUSION: The antagonist of RARα, ER-50891 could significantly attenuate ATRA's inhibitive effects on BMP 2-induced osteoblastogenesis.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , Osteogenesis/drug effects , Retinoic Acid Receptor alpha/antagonists & inhibitors , Tretinoin/antagonists & inhibitors , 3T3 Cells , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Retinoic Acid Receptor alpha/metabolism , Tretinoin/pharmacology
4.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118563, 2020 02.
Article in English | MEDLINE | ID: mdl-31666191

ABSTRACT

Skeletal muscle secretes biologically active proteins that contribute to muscle hypertrophy in response to either exercise or dietary intake. The identification of skeletal muscle-secreted proteins that induces hypertrophy can provide critical information regarding skeletal muscle health. Dietary provitamin A, ß-carotene, induces hypertrophy of the soleus muscle in mice. Here, we hypothesized that skeletal muscle produces hypertrophy-inducible secretory proteins via dietary ß-carotene. Knockdown of retinoic acid receptor (RAR) γ inhibited the ß-carotene-induced increase soleus muscle mass in mice. Using RNA sequencing, bioinformatic analyses, and literature searching, we predicted transglutaminase 2 (TG2) to be an all-trans retinoic acid (ATRA)-induced secretory protein in cultured C2C12 myotubes. Tg2 mRNA expression increased in ATRA- or ß-carotene-stimulated myotubes and in the soleus muscle of ß-carotene-treated mice. Knockdown of RARγ inhibited ß-carotene-increased mRNA expression of Tg2 in the soleus muscle. ATRA increased endogenous TG2 levels in conditioned medium from myotubes. Extracellular TG2 promoted the phosphorylation of Akt, mechanistic target of rapamycin (mTOR), and ribosomal p70 S6 kinase (p70S6K), and inhibitors of mTOR, phosphatidylinositol 3-kinase, and Src (rapamycin, LY294002, and Src I1, respectively) inhibited TG2-increased phosphorylation of mTOR and p70S6K. Furthermore, extracellular TG2 promoted protein synthesis and hypertrophy in myotubes. TG2 mutant lacking transglutaminase activity exerted the same effects as wild-type TG2. Knockdown of G protein-coupled receptor 56 (GPR56) inhibited the effects of TG2 on mTOR signaling, protein synthesis, and hypertrophy. These results indicated that TG2 expression was upregulated through ATRA-mediated RARγ and that extracellular TG2 induced myotube hypertrophy by activating mTOR signaling-mediated protein synthesis through GPR56, independent of transglutaminase activity.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Transglutaminases/metabolism , Animals , Cell Enlargement/drug effects , Cell Line , GTP-Binding Proteins/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Phosphorylation/drug effects , Protein Glutamine gamma Glutamyltransferase 2 , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha/antagonists & inhibitors , Retinoic Acid Receptor alpha/genetics , Retinoic Acid Receptor alpha/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transglutaminases/genetics , Tretinoin/pharmacology , beta Carotene/administration & dosage , beta Carotene/pharmacology , Retinoic Acid Receptor gamma
5.
Arch Pharm Res ; 42(8): 684-694, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31214877

ABSTRACT

PML/retinoic acid receptor alpha (RARα), as a hallmark of acute promyeloid leukemia (APL), is directly related to the outcome of clinical APL remedy. It is reported that arsenicals can effectively degrade PML/RARα, such as arsenic trioxide and realgar. However, the high toxicity or insolubility have hampered their clinical applications. Realgar transforming solution (RTS) was produced from realgar by bioleaching process in our lab. Previous studies demonstrated that RTS had a significant anti-cancer ability on chronic myeloid leukemia through oncoprotein degradation. The capacity of RTS on treating APL is what is focused on in this study. The results showed that RTS had a noticeable sensitivity in NB4 cell, and RTS remarkably down-regulated PML/RARα expression and induced cell differentiation. Further, RTS could accumulate PML/RARα into the nuclear bodies and then execute degradation, which could be reversed by proteasome inhibitor MG132. The results also exhibited that the reduction of RTS-induced PML/RARα expression accompanied by the elevation of ubiquitin and SUMO-1 protein expression. Finally, PML and SUMO-1 had been demonstrated to be co-localized after RTS treatment by immunofluorescence co-localization assay and immunoprecipitation assay. In conclusion, these results suggested that RTS-induced cell differentiation may attribute to the PML/RARα degradation partially through the ubiquitin-proteasome pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Promyelocytic Leukemia Protein/antagonists & inhibitors , Proteasome Endopeptidase Complex/metabolism , Retinoic Acid Receptor alpha/antagonists & inhibitors , Sulfides/pharmacology , Ubiquitins/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Promyelocytic Leukemia Protein/metabolism , Retinoic Acid Receptor alpha/metabolism , Solutions , Structure-Activity Relationship , Tumor Cells, Cultured , Ubiquitins/metabolism
6.
Cell Physiol Biochem ; 50(6): 2390-2405, 2018.
Article in English | MEDLINE | ID: mdl-30423583

ABSTRACT

BACKGROUND/AIMS: Phenotypic switching of vascular smooth muscle cells (VSMC) plays a vital role in the development of vascular diseases. All-trans retinoic acid (ATRA) is known to regulate VSMC phenotypes. However, the underlying mechanisms remain completely unknown. Here, we have investigated the probable roles and underlying mechanisms of the novel C2H2 zinc finger transcription factor ZFP580 on ATRA-induced VSMC differentiation. METHODS: VSMCs were isolated, cultured, and identified. VSMCs were infected with an adenovirus encoding ZFP580 or Ad-siRNA to silence ZFP580. The expression levels of ZFP580, SMα-actin, SM22α, SMemb, RARα, RARß, and RARγ were assayed by Q-PCR and western blot. A rat carotid artery injury model and morphometric analysis of intimal thickening were also used in this study. RESULTS: ATRA caused a significant reduction of VSMC proliferation and migration in a doseand time-dependent manner. Moreover, it promoted VSMC differentiation by enhancing expression of differentiation markers and reducing expression of dedifferentiation markers. This ATRA activity was accompanied by up-regulation of ZFP580, with concomitant increases in RARα expression. In contrast, silencing of the RARα gene or inhibiting RARα with its antagonist Ro41-5253 abrogated the ATRA-induced ZFP580 expression. Furthermore, ATRA binding to RARα induced ZFP580 expression via the PI3K/Akt and ERK pathways. Adenovirusmediated overexpression of ZFP580 promoted VSMC differentiation by enhancing expression of SM22α and SMα-actin and reducing expression of SMemb. In contrast, silencing ZFP580 dramatically reduced the expression of differentiation markers and increased expression of dedifferentiation markers. The classic rat carotid artery balloon injury model demonstrated that ZFP580 inhibited proliferation and intimal hyperplasia in vivo. CONCLUSION: The novel zinc finger transcription factor ZFP580 facilitates ATRA-induced VSMC differentiation by the RARα-mediated PI3K/Akt and ERK signaling pathways. This might represent a novel mechanism of regulation of ZFP580 by ATRA and RARα, which is critical for understanding the biological functions of retinoids during VSMC phenotypic modulation.


Subject(s)
Cell Differentiation/drug effects , Retinoic Acid Receptor alpha/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Tretinoin/pharmacology , Animals , Benzoates/pharmacology , Carotid Artery Diseases/etiology , Carotid Artery Diseases/pathology , Carotid Artery Diseases/veterinary , Cell Proliferation/drug effects , Chromans , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Retinoic Acid Receptor alpha/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
7.
Int J Oncol ; 51(3): 899-906, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28766684

ABSTRACT

Acute promyelocytic leukemia (APL) is a distinctive subtype of acute myeloid leukemia (AML) in which the hybrid protein promyelocytic leukemia protein/retinoic acid receptor α (PML/RARα) acts as a transcriptional repressor impairing the expression of genes that are critical to myeloid cell mutation. We aimed at explaining the molecular mechanism of green tea polyphenol epigallocatechin-3-gallate (EGCG) enhancement of ATRA-induced APL cell line differentiation. Tumor suppressor phosphatase and tensin homolog (PTEN) was found downregulated in NB4 cells and rescued by proteases inhibitor MG132. A significant increase of PTEN levels was found in NB4, HL-60 and THP-1 cells upon ATRA combined with EGCG treatment, paralleled by increased myeloid differentiation marker CD11b. EGCG in synergy with ATRA promote degradation of PML/RARα and restores PML expression, and increase the level of nuclear PTEN. Pretreatment of PTEN inhibitor SF1670 enhances the PI3K signaling pathway and represses NB4 cell differentiation. Moreover, the induction of PTEN attenuated the Akt phosphorylation levels, pretreatment of PI3K inhibitor LY294002 in NB4 cells, significantly augmented the cell differentiation and increased the expression of PTEN. These results therefore indicate that EGCG targets PML/RARα oncoprotein for degradation and potentiates differentiation of promyelocytic leukemia cells in combination with ATRA via PTEN.


Subject(s)
Catechin/analogs & derivatives , Leukemia, Promyelocytic, Acute/drug therapy , PTEN Phosphohydrolase/genetics , Promyelocytic Leukemia Protein/genetics , Retinoic Acid Receptor alpha/genetics , Catechin/administration & dosage , Cell Differentiation/drug effects , Chromones/administration & dosage , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Leupeptins/administration & dosage , Morpholines/administration & dosage , PTEN Phosphohydrolase/antagonists & inhibitors , Phenanthrenes/administration & dosage , Promyelocytic Leukemia Protein/antagonists & inhibitors , Proteolysis/drug effects , Retinoic Acid Receptor alpha/antagonists & inhibitors , Tretinoin/administration & dosage
8.
J Nutr Biochem ; 47: 75-85, 2017 09.
Article in English | MEDLINE | ID: mdl-28570942

ABSTRACT

This study investigated the prenatal marginal vitamin A deficiency (mVAD)-related impairment in learning and memory and the interactions between RARα, Src and NR1. Learning and memory were assessed in adult rats that were exposed to prenatal mVAD with Morris water maze. The average escape time was longer in mVAD rats, and they passed the hidden platform fewer times during the memory retention test than normal vitamin A intake (VAN) rats. The mRNA and protein levels of RARα, Src and NR1 in mVAD rats were significantly lower than those in VAN rats. RARα and Src, but not NR1, were in the same protein complex. RA treatment-induced increase in RARα, Src and NR1 expressions in mVAD neurons was much lower than that in VAN neurons. Overexpression of RARα gene in VAN neurons induced an increase in RARα, Src and NR1 expressions, while silencing of RARα gene induced a decrease in expressions of RARα and Src, but not that of of NR1. In mVAD neurons, however, overexpression of RARα did not induce an increase in NR1 expression, while silencing of RARα gene had no effect on Src and NR1 expressions. Furthermore, inhibition of Src was associated with a decrease in NR1 expression but not that of RARα. Prenatal mVAD was associated with impaired learning and memory in adult rats. It is possible that mVAD-related decrease in RARα led to a decrease in Src expression, which in turn down-regulated NR1 expression and Ca2+ influx and eventually caused learning and memory deficits.


Subject(s)
Hippocampus/metabolism , Learning Disabilities/etiology , Maternal Nutritional Physiological Phenomena , Memory Disorders/etiology , Nerve Tissue Proteins/metabolism , Retinoic Acid Receptor alpha/metabolism , Vitamin A Deficiency/physiopathology , Animals , Animals, Newborn , Cells, Cultured , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Hippocampus/pathology , Humans , Learning Disabilities/metabolism , Learning Disabilities/pathology , Learning Disabilities/prevention & control , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Memory Disorders/prevention & control , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Pregnancy , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA Interference , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Retinoic Acid Receptor alpha/agonists , Retinoic Acid Receptor alpha/antagonists & inhibitors , Retinoic Acid Receptor alpha/genetics , Severity of Illness Index , Tretinoin/therapeutic use , Vitamin A/blood , Vitamin A Deficiency/blood , Vitamin A Deficiency/diet therapy
9.
Biochim Biophys Acta Proteins Proteom ; 1865(9): 1195-1206, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28642153

ABSTRACT

Retinoic Acid Receptor alpha (RARα/NR1B1), Retinoic Acid Receptor beta (RARß/NR1B2) and Retinoic Acid Receptor gamma (RARγ/NR1B3) are transcription factors regulating gene expression in response to retinoids. Within the RAR genomic pathways, binding of RARs to coregulators is a key intermediate regulatory phase. However, ligand-dependent interactions between the wide variety of coregulators that may be present in a cell and the different RAR subtypes are largely unknown. The aim of this study is to characterize the coregulator binding profiles of RARs in the presence of the pan-agonist all-trans-Retinoic Acid (AtRA); the subtype-selective agonists Am80 (RARα), CD2314 (RARß) and BMS961 (RARγ); and the antagonist Ro415253. To this end, we used a microarray assay for coregulator-nuclear receptor interactions to assess RAR binding to 154 motifs belonging to >60 coregulators. The results revealed a high number of ligand-dependent RAR-coregulator interactions among all RAR variants, including many binding events not yet described in literature. Next, this work confirmed a greater ligand-independent activity of RARß compared to the other RAR subtypes based on both higher basal and lower ligand-driven coregulator binding. Further, several coregulator motifs showed selective binding to a specific RAR subtype. Next, this work showed that subtype-selective agonists can be successfully discriminated by using coregulator binding assays. Finally this study demonstrated the possible applications of a coregulator binding assay as a tool to discriminate between agonistic/antagonistic actions of ligands. The RAR-coregulator interactions found will be of use to direct further studies to better understand the mechanisms driving the eventual actions of retinoids.


Subject(s)
Receptors, Retinoic Acid/chemistry , Retinoic Acid Receptor alpha/chemistry , Amino Acid Motifs , Anthracenes/pharmacology , Benzoates/pharmacology , Binding Sites , Chromans , Protein Array Analysis , Protein Binding , Protein Domains , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Recombinant Proteins/metabolism , Response Elements , Retinoic Acid Receptor alpha/agonists , Retinoic Acid Receptor alpha/antagonists & inhibitors , Retinoids/pharmacology , Structure-Activity Relationship , Tetrahydronaphthalenes/pharmacology , Thiophenes/pharmacology , Tretinoin/pharmacology , Retinoic Acid Receptor gamma
10.
Arch Immunol Ther Exp (Warsz) ; 65(1): 69-81, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27412076

ABSTRACT

Activities of the retinoic acid receptor (RAR)α and RARγ are important to hematopoiesis. Here, we have investigated the effects of receptor selective agonists and antagonists on the primitive human hematopoietic cell lines KG1 and NB-4 and purified normal human hematopoietic stem cells (HSCs). Agonizing RARα (by AGN195183) was effective in driving neutrophil differentiation of NB-4 cells and this agonist synergized with a low amount (10 nM) of 1α,25-dihydroxyvitamin D3 to drive monocyte differentiation of NB-4 and KG1 cells. Treatment of cultures of human HSCs (supplemented with stem cell factor ± interleukin 3) with an antagonist of all RARs (AGN194310) or of RARα (AGN196996) prolonged the lifespan of cultures, up to 55 days, and increased the production of neutrophils and monocytes. Slowing down of cell differentiation was not observed, and instead, hematopoietic stem and progenitor cells had expanded in number. Antagonism of RARγ (by AGN205728) did not affect cultures of HSCs. Studies of CV-1 and LNCaP cells transfected with RAR expression vectors and a reporter vector revealed that RARγ and RARß are activated by sub-nM all-trans retinoic acid (EC50-0.3 nM): ~50-fold more is required for activation of RARα (EC50-16 nM). These findings further support the notion that the balance of expression and activity of RARα and RARγ are important to hematopoietic stem and progenitor cell expansion and differentiation.


Subject(s)
Hematopoietic Stem Cells/cytology , Myeloid Cells/immunology , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha/metabolism , Animals , Antigens, CD34/metabolism , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , HL-60 Cells , Haplorhini , Hematopoiesis , Humans , Neutrophils/cytology , Protein Binding , Receptors, Retinoic Acid/antagonists & inhibitors , Retinoic Acid Receptor alpha/agonists , Retinoic Acid Receptor alpha/antagonists & inhibitors , Retinoids/pharmacology , Tretinoin/chemistry , Retinoic Acid Receptor gamma
11.
Cell Biol Int ; 40(4): 456-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26818829

ABSTRACT

The regulation of vascular endothelial growth factors C (VEGF-C) and D (VEGF-D), and their receptor VEGFR3 gene and protein expression by all-trans-retinoic acid (atRA) in A549 lung cancer cells, was investigated. We showed that atRA treatment increased VEGF-C, VEGF-D, and VEGFR3 protein and mRNA contents in dose-dependent manner. atRA-mediated increase of both ligands and receptor expression correlated with the elevated level of retinoic acid receptor α (RARα) expression, while the level of another atRA receptor, peroxisome proliferator-activated receptor ß/δ (PPARß/δ), was decreased. We demonstrated that the classical counterpart of RARα, retinoid X receptor α (RXRα), was down-regulated in both cytoplasm and nucleus of A549 cells upon atRA addition. On the contrary, the nuclear quantity of another possible RARα counterpart, transcription factor Sp1, was increased after atRA treatment.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Retinoic Acid Receptor alpha/metabolism , Tretinoin/pharmacology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microscopy, Fluorescence , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Retinoic Acid Receptor alpha/antagonists & inhibitors , Retinoic Acid Receptor alpha/genetics , Signal Transduction/drug effects , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL