Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Blood ; 137(8): 1090-1103, 2021 02 25.
Article in English | MEDLINE | ID: mdl-32976550

ABSTRACT

The nuclear receptor (NR) subclass, retinoid X receptors (RXRs), exert immunomodulatory functions that control inflammation and metabolism via homodimers and heterodimers, with several other NRs, including retinoic acid receptors. IRX4204 is a novel, highly specific RXR agonist in clinical trials that potently and selectively activates RXR homodimers, but not heterodimers. In this study, in vivo IRX4204 compared favorably with FK506 in abrogating acute graft-versus-host disease (GVHD), which was associated with inhibiting allogeneic donor T-cell proliferation, reducing T-helper 1 differentiation, and promoting regulatory T-cell (Treg) generation. Recipient IRX4204 treatment reduced intestinal injury and decreased IFN-γ and TNF-α serum levels. Transcriptional analysis of donor T cells isolated from intestines of GVHD mice treated with IRX4204 revealed significant decreases in transcripts regulating proinflammatory pathways. In vitro, inducible Treg differentiation from naive CD4+ T cells was enhanced by IRX4204. In vivo, IRX4204 increased the conversion of donor Foxp3- T cells into peripheral Foxp3+ Tregs in GVHD mice. Using Foxp3 lineage-tracer mice in which both the origin and current FoxP3 expression of Tregs can be tracked, we demonstrated that IRX4204 supports Treg stability. Despite favoring Tregs and reducing Th1 differentiation, IRX4204-treated recipients maintained graft-versus-leukemia responses against both leukemia and lymphoma cells. Notably, IRX4204 reduced in vitro human T-cell proliferation and enhanced Treg generation in mixed lymphocyte reaction cultures. Collectively, these beneficial effects indicate that targeting RXRs with IRX4204 could be a novel approach to preventing acute GVHD in the clinic.


Subject(s)
Bone Marrow Transplantation , Cyclopropanes/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Leukemia Effect/drug effects , Retinoid X Receptors/agonists , Animals , Bone Marrow Transplantation/adverse effects , Drug Repositioning , Female , Graft vs Host Disease/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology
2.
Oncotarget ; 7(7): 7469-79, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26862735

ABSTRACT

Parkinson's disease (PD) is one of the most common movement disorders, and currently there is no effective treatment that can slow disease progression. Preserving and enhancing DA neuron survival is increasingly regarded as the most promising therapeutic strategy for treating PD. IRX4204 is a second generation retinoid X receptor (RXR) agonist that has no cross reactivity with retinoic acid receptors, farnesoid X receptor, liver X receptors or peroxisome proliferator-activated receptor PPARγ. We found that IRX4204 promotes the survival and maintenance of nigral dopaminergic (DA) neurons in a dose-dependent manner in primary mesencephalic cultures. Brain bioavailability studies demonstrate that IRX4204 can cross the blood brain barrier and reach the brain at nM concentration. Oral administration of IRX4204 can activate nuclear receptor Nurr1 downstream signaling in the substantia nigra (SN) andattenuate neurochemical and motor deficits in a rat model of PD. Our study suggests that IRX4204 represents a novel, potent and selective pharmacological means to activate cellular RXR-Nurr1 signaling and promote SN DA neuron survival in PD prevention and/or treatment.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/metabolism , Cyclopropanes/pharmacology , Dopaminergic Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Parkinson Disease/drug therapy , Substantia Nigra/metabolism , Trans-Activators/pharmacology , Animals , Behavior, Animal/drug effects , Blotting, Western , Brain/drug effects , Cell Membrane Permeability/drug effects , Cells, Cultured , Dopaminergic Neurons/drug effects , Immunoenzyme Techniques , Male , Neuroprotection/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/drug effects , Tandem Mass Spectrometry
3.
J Steroid Biochem Mol Biol ; 150: 46-53, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25800721

ABSTRACT

Vitamin A is known to influence post-natal bone content, with excess intake being associated with reduced bone mineral density and increased fracture risk. Despite this, the roles retinoids play in regulating osteoclastogenesis, particularly in vivo, remain unresolved. This study therefore aimed to determine the effect of loss of retinoic acid receptors (RAR)α or RARγ on bone mass (analyzed by histomorphometry and dual-energy X-ray absorptiometry) and osteoclastogenesis in mice in vivo. RARγ null mice had significantly less trabecular bone at 8 weeks of age compared to wildtype littermates. In contrast, no change in trabecular bone mass was detected in RARα null mice at this age. Further histomorphometric analysis revealed a significantly greater osteoclast surface in bones from 8-week-old RARγ null male mice. This in vivo effect was cell lineage autonomous, and was associated with increased osteoclastogenesis in vitro from hematopoietic cells obtained from 8-week-old RARγ null male mice. The use of highly selective agonists in RANKL-induced osteoclast differentiation of wild type mouse whole bone marrow cells and RAW264.7 cells in vitro showed a stronger inhibitory effect of RARγ than RARα agonists, suggesting that RARγ is a more potent inhibitor of osteoclastogenesis. Furthermore, NFAT activation was also more strongly inhibited by RARγ than RARα agonists. While RARα and RARγ antagonists did not significantly affect osteoclast numbers in vitro, larger osteoclasts were observed in cultures stimulated with the antagonists, suggesting increased osteoclast fusion. Further investigation into the effect of retinoids in vivo revealed that oral administration of 5mg/kg/day ATRA for 10 days protected against bone loss induced by granulocyte colony-stimulating factor (G-CSF) by inhibiting the pro-osteoclastogenic action of G-CSF. Collectively, our data indicates a physiological role for RARγ as a negative regulator of osteoclastogenesis in vivo and in vitro, and reveals distinct influences of RARα and RARγ in bone structure regulation.


Subject(s)
Bone Resorption/genetics , Bone and Bones/metabolism , Osteoclasts/metabolism , Receptors, Retinoic Acid/genetics , Tretinoin/pharmacology , Animals , Bone Density/drug effects , Bone Resorption/metabolism , Bone Resorption/pathology , Bone Resorption/prevention & control , Bone and Bones/drug effects , Bone and Bones/pathology , Cell Differentiation , Gene Expression Regulation, Neoplastic , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/pathology , Primary Cell Culture , RANK Ligand/genetics , RANK Ligand/metabolism , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , Tretinoin/analogs & derivatives , Retinoic Acid Receptor gamma
4.
Development ; 141(11): 2260-70, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24821986

ABSTRACT

Retinoic acid receptor gamma 2 (RARγ2) is the major RAR isoform expressed throughout the caudal axial progenitor domain in vertebrates. During a microarray screen to identify RAR targets, we identified a subset of genes that pattern caudal structures or promote axial elongation and are upregulated by increased RAR-mediated repression. Previous studies have suggested that RAR is present in the caudal domain, but is quiescent until its activation in late stage embryos terminates axial elongation. By contrast, we show here that RARγ2 is engaged in all stages of axial elongation, not solely as a terminator of axial growth. In the absence of RA, RARγ2 represses transcriptional activity in vivo and maintains the pool of caudal progenitor cells and presomitic mesoderm. In the presence of RA, RARγ2 serves as an activator, facilitating somite differentiation. Treatment with an RARγ-selective inverse agonist (NRX205099) or overexpression of dominant-negative RARγ increases the expression of posterior Hox genes and that of marker genes for presomitic mesoderm and the chordoneural hinge. Conversely, when RAR-mediated repression is reduced by overexpressing a dominant-negative co-repressor (c-SMRT), a constitutively active RAR (VP16-RARγ2), or by treatment with an RARγ-selective agonist (NRX204647), expression of caudal genes is diminished and extension of the body axis is prematurely terminated. Hence, gene repression mediated by the unliganded RARγ2-co-repressor complex constitutes a novel mechanism to regulate and facilitate the correct expression levels and spatial restriction of key genes that maintain the caudal progenitor pool during axial elongation in Xenopus embryos.


Subject(s)
Gene Expression Regulation, Developmental , Receptors, Retinoic Acid/metabolism , Animals , Apoptosis , Cell Differentiation/genetics , Co-Repressor Proteins/metabolism , Gene Expression Regulation , Genes, Dominant , Homeodomain Proteins/metabolism , Humans , Mesoderm/metabolism , Mesoderm/physiology , Nervous System/embryology , Nervous System/growth & development , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Retinoic Acid/agonists , Repressor Proteins/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , Somites/physiology , Time Factors , Xenopus Proteins/metabolism , Xenopus laevis , Retinoic Acid Receptor gamma
5.
Blood ; 122(12): 2125-34, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23814022

ABSTRACT

Graft-versus-host disease (GVHD) is a critical complication after allogeneic bone marrow transplantation. During GVHD, donor T cells are activated by host antigen-presenting cells and differentiate into T-effector cells (Teffs) that migrate to GVHD target organs. However, local environmental factors influencing Teff differentiation and migration are largely unknown. Vitamin A metabolism within the intestine produces retinoic acid, which contributes to intestinal homeostasis and tolerance induction. Here, we show that the expression and function of vitamin A-metabolizing enzymes were increased in the intestine and mesenteric lymph nodes in mice with active GVHD. Moreover, transgenic donor T cells expressing a retinoic acid receptor (RAR) response element luciferase reporter responded to increased vitamin A metabolites in GVHD-affected organs. Increasing RAR signaling accelerated GVHD lethality, whereas donor T cells expressing a dominant-negative RARα (dnRARα) showed markedly diminished lethality. The dnRARα transgenic T cells showed reduced Th1 differentiation and α4ß7 and CCR9 expression associated with poor intestinal migration, low GVHD pathology, and reduced intestinal permeability, primarily via CD4(+) T cells. The inhibition of RAR signaling augmented donor-induced Treg generation and expansion in vivo, while preserving graft-versus-leukemia effects. Together, these results suggested that reagents blunting donor T-cell RAR signaling may possess therapeutic anti-GVHD properties.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Intestines/immunology , Receptors, Retinoic Acid/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Bone Marrow Transplantation/adverse effects , Cell Differentiation/immunology , Graft vs Host Disease/mortality , Mice , Receptors, Lymphocyte Homing/metabolism , Retinoic Acid Receptor alpha , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Tretinoin/metabolism , Tretinoin/pharmacology
6.
Cancer Res ; 72(20): 5230-9, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22902413

ABSTRACT

While vitamin A has been implicated in host resistance to infectious disease, little is known about the role of vitamin A and its active metabolite, retinoic acid (RA) in host defenses against cancer. Here, we show that local RA production within the tumor microenvironment (TME) is increased up to 5-fold as compared with naïve surrounding tissue, with a commensurate increase in RA signaling to regionally infiltrating tumor-reactive T cells. Conditional disruption of RA signaling in CD8(+) T cells using a dominant negative retinoic acid receptor α (dnRARα) established that RA signaling is required for tumor-specific CD8(+) T-cell expansion/accumulation and protective antitumor immunity. In vivo analysis of antigen-specific CD8(+) T-cell responses revealed that early T-cell expansion was RA-independent; however, late T-cell expansion and clonal accumulation was suppressed strongly in the absence of RA signaling. Our findings indicate that RA function is essential for the survival of tumor-reactive CD8(+) T cells within the TME.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Survival , Tretinoin/metabolism , Tumor Microenvironment , Animals , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Signal Transduction
7.
J Exp Med ; 208(9): 1767-75, 2011 Aug 29.
Article in English | MEDLINE | ID: mdl-21859847

ABSTRACT

It is known that vitamin A and its metabolite, retinoic acid (RA), are essential for host defense. However, the mechanisms for how RA controls inflammation are incompletely understood. The findings presented in this study show that RA signaling occurs concurrent with the development of inflammation. In models of vaccination and allogeneic graft rejection, whole body imaging reveals that RA signaling is temporally and spatially restricted to the site of inflammation. Conditional ablation of RA signaling in T cells significantly interferes with CD4(+) T cell effector function, migration, and polarity. These findings provide a new perspective of the role of RA as a mediator directly controlling CD4(+) T cell differentiation and immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/physiology , Cell Movement/physiology , Immunity, Cellular/physiology , Models, Immunological , Signal Transduction/physiology , Tretinoin/immunology , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/metabolism , Immunity, Cellular/drug effects , Immunization , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Signal Transduction/drug effects , Skin Transplantation/immunology , Transplantation, Homologous , Tretinoin/metabolism , Tretinoin/pharmacology
8.
Proc Natl Acad Sci U S A ; 108(21): 8698-703, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555593

ABSTRACT

Retinoic acid receptor (RAR) signaling is required for morphogenesis of the ventral optic cup and closure of the choroid fissure, but the mechanisms by which this pathway regulates ventral eye development remain controversial and poorly understood. Although previous studies have implicated neural crest-derived periocular mesenchyme (POM) as the critical target of RA action in the eye, we show here that RAR signaling regulates choroid fissure closure in zebrafish by acting on both the ventral optic cup and the POM. We describe RAR-dependent regulation of eight genes in the neuroepithelial cells of the ventral retina and optic stalk and of six genes in the POM and show that these ventral retina/optic stalk and POM genes function independently of each other. Consequently, RAR signaling regulates ventral eye development through two independent, nonredundant mechanisms in different ocular tissues. Furthermore, the identification of two cohorts of genes implicated in ventral eye morphogenesis may help to elucidate the genetic basis of ocular coloboma in humans.


Subject(s)
Choroid/ultrastructure , Eye/growth & development , Mesoderm , Receptors, Retinoic Acid/metabolism , Signal Transduction/physiology , Animals , Choroid/metabolism , Coloboma , Embryo, Nonmammalian , Humans , Morphogenesis , Neural Crest/cytology , Optic Nerve/abnormalities , Zebrafish
9.
Nat Med ; 17(4): 454-60, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21460849

ABSTRACT

Heterotopic ossification consists of ectopic bone formation within soft tissues after surgery or trauma. It can have debilitating consequences, but there is no definitive cure. Here we show that heterotopic ossification was essentially prevented in mice receiving a nuclear retinoic acid receptor-γ (RAR-γ) agonist. Side effects were minimal, and there was no significant rebound effect. To uncover the mechanisms of these responses, we treated mouse mesenchymal stem cells with an RAR-γ agonist and transplanted them into nude mice. Whereas control cells formed ectopic bone masses, cells that had been pretreated with the RAR-γ agonist did not, suggesting that they had lost their skeletogenic potential. The cells became unresponsive to rBMP-2 treatment in vitro and showed decreases in phosphorylation of Smad1, Smad5 and Smad8 and in overall levels of Smad proteins. In addition, an RAR-γ agonist blocked heterotopic ossification in transgenic mice expressing activin receptor-like kinase-2 (ALK2) Q207D, a constitutively active form of the receptor that is related to ALK2 R206H found in individuals with fibrodysplasia ossificans progressiva. The data indicate that RAR-γ agonists are potent inhibitors of heterotopic ossification in mouse models and, thus, may also be effective against injury-induced and congenital heterotopic ossification in humans.


Subject(s)
Ossification, Heterotopic/drug therapy , Receptors, Retinoic Acid/agonists , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Chondrogenesis/drug effects , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Nude , Mice, Transgenic , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/genetics , Signal Transduction/drug effects , Retinoic Acid Receptor gamma
10.
Dev Biol ; 347(1): 195-203, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20801112

ABSTRACT

Mice deficient in growth differentiation factor 11 (GDF11) signaling display anterior transformation of axial vertebrae and truncation of caudal vertebrae. However, the in vivo molecular mechanisms by which GDF11 signaling regulates the development of the vertebral column have yet to be determined. We found that Gdf11 and Acvr2b mutants are sensitive to exogenous RA treatment on vertebral specification and caudal vertebral development. We show that diminished expression of Cyp26a1, a retinoic acid inactivating enzyme, and concomitant elevation of retinoic acid activity in the caudal region of Gdf11(-/-) embryos may account for this phenomenon. Reduced expression or function of Cyp26a1 enhanced anterior transformation of axial vertebrae in wild-type and Acvr2b mutants. Furthermore, a pan retinoic acid receptor antagonist (AGN193109) could lessen the anterior transformation phenotype and rescue the tail truncation phenotype of Gdf11(-/-) mice. Taken together, these results suggest that GDF11 signaling regulates development of caudal vertebrae and is involved in specification of axial vertebrae in part by maintaining Cyp26a1 expression, which represses retinoic acid activity in the caudal region of embryos during the somitogenesis stage.


Subject(s)
Body Patterning , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Signal Transduction , Spine/embryology , Spine/metabolism , Tretinoin/metabolism , Activin Receptors, Type II/metabolism , Animals , Body Patterning/drug effects , Body Patterning/genetics , Bone Morphogenetic Proteins/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Embryo, Mammalian/drug effects , Embryo, Mammalian/enzymology , Gene Expression Regulation, Developmental/drug effects , Growth Differentiation Factors/genetics , Mesoderm/drug effects , Mesoderm/embryology , Mesoderm/enzymology , Mice , Mutation/genetics , Retinoic Acid 4-Hydroxylase , Signal Transduction/drug effects , Somites/drug effects , Somites/embryology , Somites/enzymology , Spine/drug effects , Tail/abnormalities , Tail/drug effects , Tretinoin/pharmacology , Wnt Proteins/metabolism , Wnt3 Protein
11.
J Orthop Res ; 28(2): 271-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19725108

ABSTRACT

Heterotopic ossification (HO) consists of formation of ectopic cartilage followed by endochondral bone and is triggered by major surgeries, large wounds, and other conditions. Current therapies, including low-dose irradiation, are not always effective and do not target the skeletogenic process directly. Because chondrogenesis requires a decrease of nuclear retinoic acid receptor alpha (RARalpha) action, we reasoned that pharmacologic activation of this receptor pathway should inhibit HO. Thus, we selected the synthetic retinoid NRX195183, a potent and highly selective RARalpha-agonist, and found that it did inhibit chondrogenesis in mouse limb micromass cultures. We established a mouse HO model consisting of subcutaneous implantation of Matrigel mixed with rhBMP-2. Control mice receiving daily oral doses of vehicle (peanut oil) or retinol (a natural nonactive retinoid precursor) developed large HO-like masses by days 9-12 that displayed abundant cartilage, endochondral bone, vessels, and marrow. In contrast, formation of HO-like masses was markedly reduced in companion mice receiving daily oral doses of alpha-agonist. These ectopic masses contained sharply reduced amounts of cartilage and bone, blood vessels, and TRAP-positive osteoclasts, and expressed markedly lower levels of master chondrogenic genes including Sox9, cartilage genes such as collagen XI and X, and osteogenic genes including Runx2. The data provide proof-of-principle evidence that a pharmacological strategy involving a selective RARalpha-agonist can indeed counteract an ectopic skeletal-formation process effectively and efficiently, and could thus represent a novel preventive treatment for HO.


Subject(s)
Ossification, Heterotopic/drug therapy , Osteogenesis/drug effects , Protective Agents/administration & dosage , Receptors, Retinoic Acid/administration & dosage , Receptors, Retinoic Acid/agonists , Animals , Chondrogenesis/drug effects , Chondrogenesis/genetics , Disease Models, Animal , Gene Expression/drug effects , Mice , Ossification, Heterotopic/prevention & control , Osteogenesis/genetics , Retinoic Acid Receptor alpha , Treatment Outcome
12.
J Steroid Biochem Mol Biol ; 112(1-3): 25-31, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18761406

ABSTRACT

Here we have delineated regions of the retinoid X receptor alpha (RXRalpha) that are required for rexinoid (RXR agonist)-induced growth inhibition and apoptosis. Stable over-expression of RXRalpha in DT40 B lymphoma cells dramatically increased sensitivity to rexinoid-induced growth inhibition. By contrast, DT40 cells that over-expressed RXRalpha with a deletion of either the A/B or DNA binding domain (C domain) were resistant. We confirmed the importance of C domain integrity by point-mutating Cys(135) to Ser (C135S) to disrupt zinc-finger formation. Point mutating RXR Lys(201) to Thr and Arg(202) to Ala (KTRA) impairs RXR homodimer formation and does not affect RXR heterodimerization. When these mutated RXRs were over-expressed in DT40 cells, they failed to increase sensitivity to rexinoid. Over-expression did sensitize to growth inhibition by RAR and PPARgamma agonists. Over-expression of C135S mutated RXRalpha did not sensitize to RAR and PPARgamma agonists. Inhibitors of caspase-3 and/or caspase-9 blocked rexinoid-induced apoptosis, and activations of these caspases correlated with the ability of RXR mutants to induce cell death. These data show that the A/B and C domains of RXR and the ability of RXR to form homodimers are required for rexinoid-driven growth inhibition, caspase activation and subsequent apoptosis.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Retinoid X Receptor alpha/physiology , Amino Acid Sequence , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dimerization , Enzyme Activation , Humans , Molecular Sequence Data , PPAR gamma/agonists , PPAR gamma/metabolism , Protein Structure, Tertiary , Retinoid X Receptor alpha/agonists , Retinoid X Receptor alpha/antagonists & inhibitors , Retinoid X Receptors/agonists
13.
Development ; 135(4): 677-85, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18199582

ABSTRACT

In most animals, the gonads develop symmetrically, but most birds develop only a left ovary. A possible role for estrogen in this asymmetric ovarian development has been proposed in the chick, but the mechanism underlying this process is largely unknown. Here, we identify the molecular mechanism responsible for this ovarian asymmetry. Asymmetric PITX2 expression in the left presumptive gonad leads to the asymmetric expression of the retinoic-acid (RA)-synthesizing enzyme, RALDH2, in the right presumptive gonad. Subsequently, RA suppresses expression of the nuclear receptors Ad4BP/SF-1 and estrogen receptor alpha in the right ovarian primordium. Ad4BP/SF-1 expressed in the left ovarian primordium asymmetrically upregulates cyclin D1 to stimulate cell proliferation. These data suggest that early asymmetric expression of PITX2 leads to asymmetric ovarian development through up- or downregulation of RALDH2, Ad4BP/SF-1, estrogen receptor alpha and cyclin D1.


Subject(s)
Body Patterning , Ovary/embryology , Animals , Body Patterning/drug effects , Cell Proliferation/drug effects , Chick Embryo , Cyclin D1/genetics , Cyclin D1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Models, Biological , Ovary/cytology , Ovary/drug effects , Ovary/enzymology , Retinoic Acid 4-Hydroxylase , Sex Determination Processes , Signal Transduction/drug effects , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology , Homeobox Protein PITX2
14.
Clin Cancer Res ; 13(20): 6237-43, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17947492

ABSTRACT

PURPOSE: We evaluated the anti-inflammatory and growth-inhibitory properties of the novel rexinoid NRX194204 (4204) in vitro and then tested its ability to prevent and/or treat experimental lung and estrogen receptor (ER)-negative breast cancer in vivo. EXPERIMENTAL DESIGN: In cell culture studies, we measured the ability of 4204 to block the effects of lipopolysaccharide and induce apoptosis. For the lung cancer prevention studies, A/J mice were injected with the carcinogen vinyl carbamate and then fed 4204 (30-60 mg/kg diet) for 15 weeks, beginning 1 week after the administration of the carcinogen. For breast cancer prevention studies, mouse mammary tumor virus-neu mice were fed control diet or 4204 (20 mg/kg diet) for 50 weeks; for treatment, tumors at least 32 mm3 in size were allowed to form, and then mice were fed control diet or 4204 (60 mg/kg diet) for 4 weeks. RESULTS: Low nanomolar concentrations of 4204 blocked the ability of lipopolysaccharide and tumor necrosis factor-alpha to induce the release of nitric oxide and interleukin 6 and the degradation of IKBalpha in RAW264.7 macrophage-like cells. In the A/J mouse model of lung cancer, 4204 significantly (P < 0.05) reduced the number and size of tumors on the surface of the lungs and reduced the total tumor volume per slide by 64% to 81% compared with the control group. In mouse mammary tumor virus-neu mice, 4204 not only delayed the development of ER-negative mammary tumors in the prevention studies but also caused marked tumor regression (92%) or growth arrest (8%) in all of the mammary tumors when used therapeutically. CONCLUSIONS: The combined anti-inflammatory and anticarcinogenic actions of 4204 suggest that it is a promising new rexinoid that should be considered for future clinical trials.


Subject(s)
Breast Neoplasms/prevention & control , Fatty Acids, Unsaturated/pharmacology , Lung Neoplasms/prevention & control , Receptors, Retinoic Acid/metabolism , Tetrahydronaphthalenes/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anticarcinogenic Agents/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Carcinogens/chemistry , Fatty Acids, Unsaturated/chemistry , Humans , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Mammary Tumor Virus, Mouse/metabolism , Mice , Models, Biological , Models, Chemical , Receptors, Estrogen/metabolism , Tetrahydronaphthalenes/chemistry
15.
Transplantation ; 83(4): 375-84, 2007 Feb 27.
Article in English | MEDLINE | ID: mdl-17318068

ABSTRACT

BACKGROUND: Failure to mobilize adequate numbers of hematopoietic stem and progenitor cells (HSPC) is an important clinical problem. Since bone marrow (BM) neutrophils play a central role in HSPC mobilization, we hypothesized that granulocyte colony-stimulating factor (G-CSF)-mediated mobilization would be enhanced by further expanding the size of the BM granulocyte pool. METHODS: We tested the potential of the retinoic acid receptor alpha (RARalpha) specific agonist VTP195183, and the pan-RAR agonist all-trans retinoic acid (ATRA), to enhance G-CSF-mediated mobilization of HSPC, in two mouse strains. RESULTS: Pretreatment of mice with VTP195183 significantly increased the number of leukocytes, colony-forming cells, and early engrafting hematopoietic stem cells (HSC) mobilized in the blood in response to G-CSF. In contrast, ATRA had only a marginal effect on G-CSF-induced mobilization. HSPC mobilization synergy between VTP195183 and G-CSF occurred only when mice were preconditioned with VTP195183 prior to G-CSF. This preconditioning was shown to increase the numbers of granulocyte/macrophage progenitors in the BM. Treatment with VTP195183 and G-CSF was accompanied by enhanced levels of active neutrophil proteases in the BM extracellular fluid compared to G-CSF treatment alone. CONCLUSIONS: VTP195183 treatment increases the numbers of immature granulocyte progenitors in BM and subsequently synergizes to enhance G-CSF-mediated mobilization of HSPC. These data demonstrate a novel approach to improve G-CSF-induced mobilization by accelerating granulocyte maturation in the BM. These findings are currently being tested in a clinical trial of VTP195183 plus G-CSF for mobilization of HSPC in human patients.


Subject(s)
Cell Movement , Granulocyte Colony-Stimulating Factor/agonists , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Receptors, Retinoic Acid/agonists , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cells, Cultured , Granulocyte Colony-Stimulating Factor/metabolism , Guanosine Monophosphate/metabolism , Humans , Mice , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/enzymology , Peptide Hydrolases/metabolism , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Time Factors , Tretinoin/pharmacology
16.
J Cell Physiol ; 209(3): 854-65, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16972258

ABSTRACT

N-(4-hydroxyphenyl)retinamide (4HPR, fenretinide), a retinoic acid (RA) derivative and a potential cancer preventive agent, is known to exert its chemotherapeutic effects in cancer cells through induction of apoptosis. Earlier work from our laboratory has shown that relatively low concentrations of 4HPR induce neuronal differentiation of cultured human retinal pigment epithelial (ARPE-19) cells (Chen et al., 2003, J Neurochem 84:972-981). However, at higher concentrations of 4HPR, these cells showed morphological changes including cell shrinkage and cell death. Here we demonstrate that ARPE-19 cells treated with 4HPR exhibit a dose- and time-dependent induction of apoptosis as evidenced by morphological changes, mono- and oligonucleosome generation, and increased activity of caspases 2 and 3. The 4HPR-induced apoptosis as well as the activation of caspases 2 and 3 were blocked by both retinoic acid receptors (RAR) pan-antagonists, AGN193109 and AGN194310, and by an RARalpha-specific antagonist AGN194301. 4HPR treatment also increased reactive oxygen species (ROS) generation in ARPE-19 cells in a time-dependent manner as determined from the oxidation of 2',7'-dichlorofluorescin. In addition, the increase in the expression of heme oxygenase-1 (HO-1), a stress response protein, and the growth arrest and DNA damage-inducible transcription factor 153 (Gadd153) in response to the ROS generation were also blocked by these receptor antagonists. Pyrrolidine dithiocarbamate (PDTC), a free-radical scavenger, inhibited 4HPR-induced ROS generation, the expression of its downstream mediator, Gadd153, and apoptosis in the pretreated cells. Therefore, our results, clearly demonstrate that 4HPR induces apoptosis in ARPE-19 cells and that RARs mediate this process by regulating ROS generation as well as the expression of Gadd153 and HO-1.


Subject(s)
Apoptosis/physiology , Epithelial Cells , Fenretinide/pharmacology , Heme Oxygenase-1/metabolism , Pigment Epithelium of Eye/cytology , Reactive Oxygen Species/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factor CHOP/metabolism , Anticarcinogenic Agents/pharmacology , Antioxidants/metabolism , Caspase 2/genetics , Caspase 2/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Heme Oxygenase-1/genetics , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Transcription Factor CHOP/genetics
17.
Dev Biol ; 291(1): 96-109, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16423341

ABSTRACT

Retinoic acid is clearly important for the development of the heart. In this paper, we provide evidence that retinoic acid is essential for multiple aspects of cardiogenesis in Xenopus by examining embryos that have been exposed to retinoic acid receptor antagonists. Early in cardiogenesis, retinoic acid alters the expression of key genes in the lateral plate mesoderm including Nkx2.5 and HAND1, indicating that early patterning of the lateral plate mesoderm is, in part, controlled by retinoic acid. We found that, in Xenopus, the transition of the heart from a sheet of cells to a tube required retinoic acid signaling. The requirement for retinoic acid signaling was determined to take place during a narrow window of time between embryonic stages 14 and 18, well before heart tube closure. At the highest doses used, the lateral fields of myocardium fail to fuse, intermediate doses lead to a fusion of the two sides but failure to form a tube, and embryos exposed to lower concentrations of antagonist form a heart tube that failed to complete all the landmark changes that characterize looping. The myocardial phenotypes observed when exposed to the retinoic acid antagonist resemble the myocardium from earlier stages of cardiogenesis, although precocious expression of cardiac differentiation markers was not seen. The morphology of individual cells within the myocardium appeared immature, closely resembling the shape and size of cells at earlier stages of development. However, the failures in morphogenesis are not merely a slowing of development because, even when allowed to develop through stage 40, the heart tubes did not close when embryos were exposed to high levels of antagonist. Indeed, some aspects of left-right asymmetry also remained even in hearts that never formed a tube. These results demonstrate that components of the retinoic acid signaling pathway are necessary for the progression of cardiac morphogenesis in Xenopus.


Subject(s)
GATA4 Transcription Factor/physiology , Heart/embryology , Homeodomain Proteins/physiology , Transcription Factors/physiology , Tretinoin/physiology , Xenopus Proteins/physiology , Animals , Body Patterning , Embryo, Nonmammalian/metabolism , Female , GATA4 Transcription Factor/biosynthesis , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/biosynthesis , Mesoderm/physiology , Myocardium/metabolism , Signal Transduction , Transcription Factors/biosynthesis , Tretinoin/antagonists & inhibitors , Xenopus Proteins/biosynthesis , Xenopus laevis
18.
J Cell Biochem ; 97(2): 327-50, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16178010

ABSTRACT

All-trans retinoic acid and 9-cis-retinoic acid stimulate the activity of steroid sulfatase in HL60 acute myeloid leukemia cells in a concentration- and time-dependent manner. Neither of these 'natural retinoids' augmented steroid sulfatase activity in a HL60 sub-line that expresses a dominant-negative retinoic acid receptor alpha (RARalpha). Experiments with synthetic RAR and RXR agonists and antagonists suggest that RARalpha/RXR heterodimers play a role in the retinoid-stimulated increase in steroid sulfatase activity. The retinoid-driven increase in steroid sulfatase activity was attenuated by inhibition of phospholipase D (PLD), but not by inhibitors of phospholipase C. Experiments with inhibitors of protein kinase C (PKC) show that PKCalpha and PKCdelta play an important role in modulating the retinoid-stimulation of steroid sulfatase activity in HL60 cells. Furthermore, we show that pharmacological inhibition of the RAF-1 and ERK MAP kinases blocked the retinoid-stimulated increase in steroid sulfatase activity in HL60 cells and, by contrast, inhibition of the p38-MAP kinase or JNK-MAP kinase had no effect. Pharmacological inhibitors of the phosphatidylinositol 3-kinase, Akt, and PDK-1 also abrogated the retinoid-stimulated increase in steroid sulfatase activity in HL60 cells. These results show that crosstalk between the retinoid-stimulated genomic and non-genomic pathways is necessary to increase steroid sulfatase activity in HL60 cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Leukemia, Myeloid/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Retinoic Acid/metabolism , Steryl-Sulfatase/metabolism , Tretinoin/metabolism , Tretinoin/physiology , Cells, Cultured , HL-60 Cells , Humans , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/physiology , Phospholipase D/metabolism , Protein Kinases/metabolism , Receptors, Retinoic Acid/physiology , Retinoic Acid Receptor alpha , Retinoid X Receptors/metabolism , Transfection , Tretinoin/agonists , Tretinoin/antagonists & inhibitors
19.
J Immunol ; 175(12): 7916-29, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16339527

ABSTRACT

Vitamin A affects many aspects of T lymphocyte development and function. The vitamin A metabolites all-trans- and 9-cis-retinoic acid regulate gene expression by binding to the retinoic acid receptor (RAR), while 9-cis-retinoic acid also binds to the retinoid X receptor (RXR). Naive DO11.10 T lymphocytes expressed mRNA and protein for RAR-alpha, RXR-alpha, and RXR-beta. DNA microarray analysis was used to identify RXR-responsive genes in naive DO11.10 T lymphocytes treated with the RXR agonist AGN194204. A total of 128 genes was differentially expressed, including 16 (15%) involved in cell growth or apoptosis. Among these was Bcl2a1, an antiapoptotic Bcl2 family member. Quantitative real-time PCR analysis confirmed this finding and demonstrated that Bcl2a1 mRNA expression was significantly greater in nonapoptotic than in apoptotic T lymphocytes. The RXR agonist 9-cis-retinoic acid also increased Bcl2a1 expression, although all-trans-retinoic acid and ligands for other RXR partner receptors did not. Treatment with AGN194204 and 9-cis-retinoic acid significantly decreased apoptosis measured by annexin V staining but did not affect expression of Bcl2 and Bcl-xL. Bcl2a1 promoter activity was examined using a luciferase promoter construct. Both AGN194204 and 9-cis-retinoic acid significantly increased luciferase activity. In summary, these data demonstrate that RXR agonists increase Bcl2a1 promoter activity and increase expression of Bcl2a1 in naive T lymphocytes but do not affect Bcl2 and Bcl-xL expression in naive T lymphocytes. Thus, this effect on Bcl2a1 expression may account for the decreased apoptosis seen in naive T lymphocytes treated with RXR agonists.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Retinoid X Receptors/agonists , T-Lymphocytes/cytology , Alitretinoin , Animals , Apoptosis/genetics , Cells, Cultured , Fatty Acids, Unsaturated/pharmacology , Gene Expression Profiling , Gene Expression Regulation/immunology , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , T-Lymphocytes/drug effects , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology , bcl-X Protein/genetics
20.
J Biol Chem ; 280(46): 38317-27, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16179348

ABSTRACT

Agonists for the nuclear receptor peroxisomal proliferator-activated receptor-gamma (PPARgamma) and its heterodimeric partner, retinoid X receptor (RXR), are effective agents for the treatment of type 2 diabetes. To gain insight into the antidiabetic action of these compounds, we treated female Zucker diabetic rats (ZFF) with AGN194204, which we show to be a homodimer-specific RXR agonist, or the PPARgamma agonist, troglitazone. Hyperinsulinemic-euglycemic clamps in ZFF showed that troglitazone and AGN194204 reduced basal endogenous glucose production (EGP) approximately 30% and doubled the insulin suppression of EGP. AGN194204 had no effect on peripheral glucose utilization, whereas troglitazone increased insulin-stimulated glucose utilization by 50%, glucose uptake into skeletal muscle by 85%, and de novo skeletal muscle glycogen synthesis by 300%. Troglitazone increased skeletal muscle Irs-1 and phospho-Akt levels following in vivo insulin treatment, whereas AGN194204 increased hepatic Irs-2 and insulin stimulated phospho-Akt in liver. Gene profiles of AGN194204-treated mouse liver analyzed by Ingenuity Pathway Analysis identified increases in fatty acid synthetic genes, including Srebp-1 and fatty acid synthase, a pathway previously shown to be induced by RXR agonists. A network of down-regulated genes containing Foxa2, Foxa3, and G-protein subunits was identified, and decreases in these mRNA levels were confirmed by quantitative reverse transcription-PCR. Treatment of HepG2 cells with AGN194204 resulted in inhibition of glucagon-stimulated cAMP accumulation suggesting the G-protein down-regulation may provide an additional mechanism for hepatic insulin sensitization by RXR. These studies demonstrate distinct molecular events lead to insulin sensitization by high affinity RXR and PPARgamma agonists.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental , Fatty Acids, Unsaturated/pharmacology , PPAR gamma/agonists , PPAR gamma/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/metabolism , Tetrahydronaphthalenes/pharmacology , Troglitazone/pharmacology , Animals , Cell Line , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Down-Regulation/drug effects , Female , Rats , Rats, Zucker
SELECTION OF CITATIONS
SEARCH DETAIL
...