Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Genet ; 27(1): 74-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11138002

ABSTRACT

Mutations or rearrangements in the gene encoding the receptor tyrosine kinase RET result in Hirschsprung disease, cancer and renal malformations. The standard model of renal development involves reciprocal signaling between the ureteric bud epithelium, inducing metanephric mesenchyme to differentiate into nephrons, and metanephric mesenchyme, inducing the ureteric bud to grow and branch. RET and GDNF (a RET ligand) are essential mediators of these epithelial-mesenchymal interactions. Vitamin A deficiency has been associated with widespread embryonic abnormalities, including renal malformations. The vitamin A signal is transduced by nuclear retinoic acid receptors (RARs). We previously showed that two RAR genes, Rara and Rarb2, were colocalized in stromal mesenchyme, a third renal cell type, where their deletion led to altered stromal cell patterning, impaired ureteric bud growth and downregulation of Ret in the ureteric bud. Here we demonstrate that forced expression of Ret in mice deficient for both Rara and Rarb2 (Rara(-/-)Rarb2(-/-)) genetically rescues renal development, restoring ureteric bud growth and stromal cell patterning. Our studies indicate the presence of a new reciprocal signaling loop between the ureteric bud epithelium and the stromal mesenchyme, dependent on Ret and vitamin A. In the first part of the loop, vitamin-A-dependent signals secreted by stromal cells control Ret expression in the ureteric bud. In the second part of the loop, ureteric bud signals dependent on Ret control stromal cell patterning.


Subject(s)
Drosophila Proteins , Epithelium/drug effects , Kidney/embryology , Mesoderm/drug effects , Vitamin A/pharmacology , Animals , Epithelium/metabolism , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors , In Situ Hybridization , Kidney/abnormalities , Kidney/drug effects , Kidney/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , Morphogenesis/drug effects , Mutation , Organ Culture Techniques , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ret , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases , Signal Transduction/drug effects , Vitamin A/administration & dosage , Vitamin A/genetics , Vitamin A Deficiency/genetics , Vitamin A Deficiency/physiopathology
2.
Arch Biochem Biophys ; 477(1): 163-74, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18585997

ABSTRACT

We previously identified NEDD9 (RAINB2/HEF1/Cas-L) as a new downstream target of all-trans retinoic acid (atRA) and its receptors in the human neuroblastoma cell line, SH-SY5Y [R.A. Merrill, A.W.-M. See, M.L. Wertheim, M. Clagett-Dame, Dev. Dyn. 231 (2004) 564-575; R.A. Merrill, J.M. Ahrens, M.E. Kaiser, K.S. Federhart, V.Y. Poon, M. Clagett-Dame, Biol. Chem. 385 (2004) 605-614]. We now provide functional evidence that NEDD9 is directly regulated by atRA through a complex retinoic acid response element (RARE) located in the NEDD9 proximal promoter and consisting of four conserved half-sites separated by 1, 5, and 1 intervening base pairs. We show that a region of the human NEDD9 promoter from -1670 to +15 is sufficient to confer atRA-responsiveness and that a complex RARE located from -475 to -445 is necessary for this effect. While mutation of any one half-site does not eliminate complex formation in electrophoretic mobility shift assays (EMSA); these same mutations, when tested in transient transfection assays, markedly decrease atRA-responsiveness. Finally, chromatin immunoprecipitation (ChIP) assays demonstrate that RAR and RXR are bound to the RARE in cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation , Neurites/physiology , Phosphoproteins/genetics , Response Elements , Tretinoin/physiology , Adaptor Proteins, Signal Transducing/physiology , Base Sequence , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line , Chromatin Immunoprecipitation , DNA Primers , Electrophoretic Mobility Shift Assay , Exons , Humans , Neurites/drug effects , Neurites/metabolism , Phosphoproteins/physiology , RNA, Messenger/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Up-Regulation
3.
Brain Res ; 1632: 141-55, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26683084

ABSTRACT

NEDD9 (neural precursor cell expressed, developmentally down-regulated 9) is a member of the CAS (Crk-associated substrate) family of scaffolding proteins that regulate cell adhesion and migration. A Nedd9 knock-out/lacZ knock-in mouse (Nedd9(-/)(-)) was developed in order to study Nedd9 expression and function in the nervous system. Herein we show that NEDD9 is expressed in the adult brain and is prominently expressed in the hippocampus. Behavioral testing uncovered functional deficits in Nedd9(-)(/)(-) mice. In the Morris water maze test, Nedd9(-)(/)(-) mice showed deficits in both the ability to learn the task as well as in their ability to recall the platform location. There was no change in the gross morphology of the hippocampus, and stereological analysis of BrdU-labeled newly formed hippocampal cells suggested that this defect is not secondary to altered neurogenesis. However, analysis of the hippocampus revealed extensive loss of dendritic spine density in both the dentate gyrus (DG) and CA1 regions. Spine loss occurred equally across all branch orders and regions of the dendrite. Analysis of spine density in Nedd9(-)(/)(-) mice at 1.5, 6 and 10 months revealed an age-dependent spine loss. This work shows that NEDD9 is required for the maintenance of dendritic spines in the hippocampus, and suggests it could play a role in learning and memory.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Cognition Disorders/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Animals , Cognition Disorders/pathology , Dendritic Spines/pathology , Female , Gene Knock-In Techniques , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Biochim Biophys Acta ; 986(2): 271-80, 1989 Nov 27.
Article in English | MEDLINE | ID: mdl-2531613

ABSTRACT

A ligand affinity matrix has been developed and utilized to purify the dopamine D2 receptor approx. 2100 fold from bovine striatal membranes. 3-[2-Aminoethyl]-8-[3-(4-fluorobenzoyl)propyl]-4-oxo-1-phenyl-1,3,8- triazaspiro[4.5]decan-4-one (AES) was synthesized and used to prepare the affinity matrix by coupling to epoxy-activated Sepharose 6B (AES-Sepharose). AES (Ki approximately 1.7 nM) is similar in potency to the parent compound, spiperone (Ki approximately 0.8 nM), in competing for [3H]spiperone-binding activity. AES has no significant potency in competing for the dopamine D1 receptor as assessed by competition for [3H]SCH23390 binding (Ki greater than 1 microM). Covalent photoaffinity labeling of the dopamine D2 receptor in bovine striatal membranes with N-(p-azido-m-[125I]iodophenethyl)spiperone [( 125I]N3-NAPS) was prevented by AES at nanomolar concentrations. The dopamine D2 receptor was solubilized from bovine striatal membranes using 0.25% cholate in the presence of high ionic strength, followed by precipitation and subsequent treatment with 0.5% digitonin. Nearly 100% of the [3H]spiperone-binding activity in the cholate-digitonin solubilized preparation was absorbed at a receptor-to-resin ratio of 2:1 (v/v). Dopamine D2 receptor was eluted from the affinity resin using a competing dopaminergic antagonist molecule, haloperidol. Recovery of dopamine D2 receptor activity from the affinity matrix was approx. 9% of the activity adsorbed to the resin. The [3H]spiperone-binding activity in AES-Sepharose affinity purified preparations is saturable and of high affinity (0.2 nM). Affinity-purified preparations maintain the ligand-binding characteristics of a dopamine D2 receptor as assessed by agonist and antagonist competition for [3H]spiperone binding.


Subject(s)
Chromatography, Affinity , Corpus Striatum/analysis , Receptors, Dopamine/isolation & purification , Affinity Labels , Animals , Benzazepines/metabolism , Binding, Competitive , Cattle , Cell Membrane/analysis , Cholic Acid , Cholic Acids , Digitonin , Molecular Structure , Photochemistry , Receptors, Dopamine/metabolism , Receptors, Dopamine D2 , Sepharose , Spiperone/analogs & derivatives , Spiperone/chemical synthesis , Spiperone/metabolism
5.
Crit Rev Eukaryot Gene Expr ; 7(4): 299-342, 1997.
Article in English | MEDLINE | ID: mdl-9607169

ABSTRACT

The discovery and development of information surrounding the retinoic acid receptors (RAR and RXR) has ushered in a new era in understanding the molecular mechanism of action of vitamin A in embryonic development and cellular differentiation. The mechanisms involved in the regulation of gene expression by the retinoids is at least partially known and involves binding of the RAR and RXR to retinoic acid response elements. Additional factors, including coregulatory proteins, associated regulatory elements, and cell-specific factors, may also be involved in determining the specificity of retinoid-regulation of gene expression during development. During embryogenesis, retinoids are required for the development of the posterior hindbrain and its associated structures, as well as for the survival and differentiation of certain classes of neurons and neural crest cell derivatives. At least some of the effects of retinoid on hindbrain development are related to the regulation of Hox gene expression. Additional retinoid-regulated genes have been implicated in nervous system development, and the manner in which they lead to phenotypic changes during embryogenesis remains to be determined.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Nervous System/embryology , Nervous System/growth & development , Retinoids/pharmacology , Animals , Humans , Nervous System/drug effects , Retinoids/metabolism
6.
Neurosci Lett ; 174(2): 205-8, 1994 Jun 20.
Article in English | MEDLINE | ID: mdl-7970180

ABSTRACT

The expression of five muscarinic receptor mRNAs (m1, m2, m3, m4, m5) was examined in neostriatum, hippocampus and frontal cortex of 3, 18, and 33 month old rats. Transcripts for ml (3.0 kb), m2 (6.2 kb), m3 (4.5 kb) and m4 (3.3 kb) were detected in all brain regions studied. A 6.0 kb transcript for the m5 muscarinic receptor was observed in hippocampus and neostriatum, but was not detected in frontal cortex. Age-related changes in muscarinic receptor transcript expression were restricted to the neostriatum, where m3 and m4 mRNAs were decreased at both 18 and 33 months of age. The reduction in transcripts of m3 and m4 receptors may contribute to changes in cholinergic system function with age.


Subject(s)
Aging/metabolism , Brain Chemistry/physiology , Brain/growth & development , Receptors, Muscarinic/biosynthesis , Animals , Autoradiography , Blotting, Northern , In Situ Hybridization , Male , Oligonucleotide Probes , Rats , Rats, Inbred BN , Rats, Inbred F344
7.
Exp Biol Med (Maywood) ; 226(8): 766-75, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11520943

ABSTRACT

Both nerve growth factor (NGF) and neurotrophin-3 (NT-3) are necessary for the survival of embryonic sympathetic neurons in vivo. All-trans retinoic acid (atRA) has been shown to promote neurite outgrowth and long-term survival of chick embryonic sympathetic neurons cultured in the presence of NGF. The present study shows that atRA can also potentiate the survival and neurite outgrowth-promoting activities of NT-3. This was accomplished by enhancing the survival of existing neurons, as cell proliferation was unaffected by exposure to atRA. atRA also enhanced neurite outgrowth of the NT-3-treated cells; however, the neurites appeared thicker and less branched than cells treated with atRA in combination with NGF. Using a quantitative PCR assay, trkA and p75(NTR) mRNAs, but not trkC mRNA, were increased ( approximately 1.5- to 2-fold) after 72 and 48 hr of exposure of the cultures to atRA, respectively. The atRA-induced increase in trkA mRNA may play a role in the enhanced survival of neurons cultured in the presence of either NGF or NT-3, as both neurotrophins have been shown to signal through this receptor. The time course of these mRNA changes would indicate that atRA does not regulate the neurotrophin receptor mRNA directly, rather, intervening gene transcription is required. Thus, during development, atRA may play a role in fine-tuning embryonic responsiveness to both NT-3 and NGF.


Subject(s)
Neurons/cytology , Neurons/metabolism , Neurotrophin 3/therapeutic use , Tretinoin/therapeutic use , Animals , Cell Division/drug effects , Cell Survival/drug effects , Chick Embryo , Drug Interactions , Keratolytic Agents/therapeutic use , Nerve Growth Factor/pharmacology , Polymerase Chain Reaction/methods , RNA, Messenger/metabolism , Receptor, Nerve Growth Factor , Receptor, trkA/biosynthesis , Receptor, trkC/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Time Factors
8.
Anticancer Res ; 21(6A): 3839-44, 2001.
Article in English | MEDLINE | ID: mdl-11911255

ABSTRACT

The antitumor effects of N-(4-hydroxyphenyl)retinamide (4-HPR), and its stable C-linked analog, 4-hydroxybenzylretinone (4-HBR) on the regression of established 7,12-dimethylbenz(a)anthracene(DMBA)-induced rat mammary tumors were compared. 4-HBR is a stable and nonhydroyzable derivative which cannot be converted in vivo to retinoic acid (RA). The results indicate that 4-HBR decreased mammary tumor volumes to the same extent as equimolar concentration (2 mmol/kg diet) of 4-HPR (-45% for 4-HBR vs. -42% for 4-HPR, p<0.01). Both 4-HPR and 4-HBR bind very poorly to nuclear retinoid receptors RARs and RXRs. The similarity of physicochemical properties of 4-HPR and 4-HBR as well as their equal antitumor potency suggests that 4-HPR like 4-HBR, is acting directly rather than through hydrolysis to free RA. Treatment with 4-HPR caused an almost 65% decrease in serum retinol levels. These results suggest that 4-HBR may have a significant chemotherapeutic advantage over 4-HPR, as the nonhydrolyzable analog may not cause night blindness which occurs as a significant side effect of 4-HPR usage.


Subject(s)
Antineoplastic Agents/pharmacology , Fenretinide/analogs & derivatives , Mammary Neoplasms, Experimental/drug therapy , Vitamin A/pharmacology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Anticarcinogenic Agents/metabolism , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/metabolism , Carcinogens , Female , Fenretinide/metabolism , Fenretinide/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/metabolism , Vitamin A/analogs & derivatives , Vitamin A/blood , Vitamin A/metabolism
9.
Anticancer Res ; 16(2): 757-63, 1996.
Article in English | MEDLINE | ID: mdl-8687125

ABSTRACT

The O-glucuronide analog of N-(4-hydroxyphenyl)retinamide (4-HPROG) has shown a greater chemopreventive activity than the parent N-(4-hydroxyphenyl)retinamide (4-HPR). However, this compound is relatively unstable. In order to improve stability and efficacy, we have prepared a number of stable C-linked analogs of 4-HPROG (C-phenyl and C-benzyl glucuronosyl, glucosyl, and xylosyl analogs). These analogs are stable toward acid hydrolysis and the glucuronosyl analogs resist the actions of beta-glucuronidase. The analogs were prescreened for their antiproliferative potential in vitro using cultured human MCF-7 breast cancer cells. Selected analogs were then evaluated for their ability to inhibit the development and growth of tumors in the 7,12-dimethylbenzanthracene-induced rat mammary tumor model. Although the stable C-linked analogs bound poorly to the nuclear retinoic acid receptors, many showed more potency than the less stable 4-HPROG in inhibiting tumor incidence and multiplicity in vivo. The glucuronide/glucoside analogs are more potent than the xylosides, and the C-benzyl more effective than the C-phenyl analogs. The higher potency of at least two C-linked analogs (retinamidobenzyl glucuronide and retinamidobenzyl glucose) suggests that these analogs may have a chemopreventive advantage over the parent retinamide and its natural O-glucuronide.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Fenretinide/analogs & derivatives , Glucuronates/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Anticarcinogenic Agents/chemistry , Breast Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Female , Fenretinide/chemistry , Fenretinide/therapeutic use , Glucuronates/chemistry , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/prevention & control , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
10.
Anticancer Res ; 19(2A): 999-1004, 1999.
Article in English | MEDLINE | ID: mdl-10368645

ABSTRACT

The long term chemopreventive effects of the N-(4-hydroxyphenyl) retinamide-O-glucuronide (4-HPROG), and its stable C-linked benzyl glucuronide analog, retinamidobenzyl glucuronide (4-HPRCG) on the growth and development of 7,12-dimethylbenz[a]anthracene-induced mammary tumors were compared. The retinamidobenzyl glucuronide is stable toward acid hydrolysis and resists the actions of beta-glucuronidase. The results indicate that the C-linked glucuronide analog, 4-HPRCG has a greater chemopreventive potency than an equimolar concentration of 4-HPROG. Tumor latency was 15% longer in rats fed 2 mmol/kg diet of 4-HPRCG as compared to 4-HPROG. At 80 days post DMBA-intubation, tumor incidence was 57% and 27% in the 4-HPROG and 4-HPRCG treated rats, respectively. Tumor multiplicity was also markedly decreased in the 4-HPRCG treated rats. At 80 days post DMBA intubation the control rats had an average of 1.43 tumors/rat compared to 0.71 and 0.36 tumors/rat in the 4-HPROG and 4-HPRCG respectively. The higher potency and low toxicity of 4-HPRCG suggest that this stable analog may have an in vivo chemopreventive advantage over its analog, 4-HPROG. The results also demonstrated that these glucuronide analogs do not bind effectively in vitro either to the nuclear retinoid receptors or to the cellular retinoid binding proteins. Regardless of the mode of action of these retinoids, they are clearly effective chemopreventive agents.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Fenretinide/analogs & derivatives , Glucuronates/therapeutic use , Mammary Neoplasms, Experimental/prevention & control , Retinoids/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Female , Fenretinide/therapeutic use , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Rats , Rats, Sprague-Dawley
12.
Dev Neurobiol ; 68(13): 1441-53, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18726912

ABSTRACT

Neuron navigator 2 (Nav2) was first identified as an all-trans retinoic acid (atRA)-responsive gene in human neuroblastoma cells (retinoic acid-induced in neuroblastoma 1, RAINB1) that extend neurites after exposure to atRA. It is structurally related to the Caenorhabditis elegans unc-53 gene that is required for cell migration and axonal outgrowth. To gain insight into NAV2 function, the full-length human protein was expressed in C. elegans unc-53 mutants under the control of a mechanosensory neuron promoter. Transgene expression of NAV2 rescued the defects in unc-53 mutant mechanosensory neuron elongation, indicating that Nav2 is an ortholog of unc-53. Using a loss-of-function approach, we also show that Nav2 induction is essential for atRA to induce neurite outgrowth in SH-SY5Y cells. The NAV2 protein is located both in the cell body and along the length of the growing neurites of SH-SY5Y cells in a pattern that closely mimics that of neurofilament and microtubule proteins. Transfection of Nav2 deletion constructs in Cos-1 cells reveals a region of the protein (aa 837-1065) that directs localization with the microtubule cytoskeleton. Collectively, this work supports a role for NAV2 in neurite outgrowth and axonal elongation and suggests this protein may act by facilitating interactions between microtubules and other proteins such as neurofilaments that are key players in the formation and stability of growing neurites.


Subject(s)
Axons/physiology , Gene Expression Regulation/physiology , Nerve Growth Factors/physiology , Neurites/physiology , Neurons/cytology , Animals , Animals, Genetically Modified , Axons/drug effects , Caenorhabditis elegans , Caenorhabditis elegans Proteins/physiology , Cell Line , Chlorocebus aethiops , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Humans , Microfilament Proteins/physiology , Mutation/physiology , Nerve Growth Factors/genetics , Neurites/drug effects , Neuroblastoma , Neurofilament Proteins/metabolism , Neurons/drug effects , RNA Interference/physiology , Time Factors , Transfection , Tretinoin/pharmacology , Tubulin/metabolism
13.
Osteoporos Int ; 17(5): 704-15, 2006.
Article in English | MEDLINE | ID: mdl-16491322

ABSTRACT

INTRODUCTION: 2-Methylene-19-nor-(20S)-1alpha,25-dihydroxyvitamin D3 (2MD) is a new analog of 1alpha,25-dihydroxyvitamin D3 (1,25-(OH)2D3) that has unique properties (distinct from 1alpha,25-dihydroxyvitamin D3) in stimulating osteoblasts to form bone in culture. This analog has now been extensively tested in aged ovariectomized female rats maintained on a diet adequate in calcium and phosphorus. METHODS: Retired female rats obtained from Sprague-Dawley were ovariectomized, and were either dosed with vehicle or 2MD at 5-7 ng/kg body weight each day. RESULTS: A marked increase in total bone mass resulted during the 28-week study. This increase in bone mass resulted from an increase in both cortical and trabecular bone, with increases to the order of 25% in the cancellous bone. Histomorphometry revealed that 2MD increased bone mass primarily by increasing bone formation. It also revealed little or no effect on bone resorption. The resulting bone is of high quality revealed by histology and biomechanical testing. CONCLUSION: Throughout the study, serum calcium remained within the normal range and thus 2MD shows great promise for the treatment of bone diseases characterized by bone loss, including osteoporosis.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/drug therapy , Calcitriol/analogs & derivatives , Osteogenesis/drug effects , Osteoporosis/drug therapy , Anabolic Agents/therapeutic use , Analysis of Variance , Animals , Bone Density/drug effects , Calcitriol/therapeutic use , Calcium/blood , Female , Rats , Rats, Sprague-Dawley
14.
Arch Biochem Biophys ; 274(1): 145-54, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2528326

ABSTRACT

The glycoprotein nature of the binding subunit of the dopamine D2 receptor in rat striatum has been examined by photoaffinity labeling receptor preparations with N-(p-azido-m-[125I]iodophenethyl)spiperone followed by treatment of crude membrane receptor or receptor fractions isolated from sodium dodecyl sulfate (SDS) polyacrylamide gels with endo- and exoglycosidases. The major photoaffinity labeled protein migrates as a heterogeneous species on 10% SDS polyacrylamide gels and ranges from 130,000 to 75,000 relative molecular mass (Mr). This heterogeneity can be explained by glycosylation of the receptor by complex-type N-linked oligosaccharides. Three fractions of labeled receptor were isolated from SDS polyacrylamide gels over a range of 130,000 to 75,000 Mr; after digestion with peptide-N4-[N-acetyl-beta-glucosaminyl] asparagine amidase, all fractions yielded a single peptide approximately 40,000 Mr. Treatment of photoaffinity labeled membranes with alpha-mannosidase was without effect. The dopamine D2 receptor appears to contain substantial amounts of sialic acid as treatment of photoaffinity labeled membranes with neuraminidase increased the receptor mobility on SDS polyacrylamide gels to a species of 50,000-54,000 Mr. Treatment of the receptor with neuraminidase followed by endo-alpha-N-acetylgalactosaminidase did not change the electrophoretic migration pattern from that seen after neuraminidase treatment alone, suggesting that the binding peptide contains no serine- or threonine-linked oligosaccharides. A smaller binding peptide of approximately 31,000 Mr is also apparent in crude photoaffinity labeled membranes. This material also contains N-linked oligosaccharide. Complete removal of N-linked oligosaccharide from the dopamine D2 receptor did not change the rank order potency of agonist and antagonist compounds to compete for [3H]spiperone binding to crude membrane fractions. The dopamine D2 receptor represents a highly glycosylated neural receptor.


Subject(s)
Corpus Striatum/metabolism , Membrane Glycoproteins/metabolism , Oligosaccharides , Receptors, Dopamine/metabolism , Affinity Labels/metabolism , Animals , Autoradiography , Azides/metabolism , Cell Membrane/metabolism , Glycosylation , Iodine Radioisotopes , Kinetics , Male , Membrane Glycoproteins/isolation & purification , Molecular Weight , Oligosaccharides/isolation & purification , Rats , Receptors, Dopamine/isolation & purification , Receptors, Dopamine D2 , Spiperone/analogs & derivatives , Spiperone/metabolism
15.
Arch Biochem Biophys ; 319(2): 457-63, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7786028

ABSTRACT

Two families of nuclear retinoid receptors, retinoic acid receptor and retinoid X receptor (RAR and RXR respectively), and a family of cellular retinoic acid-binding proteins (CRABPI and II) participate in the retinoic acid (RA) signaling pathway. The presence and function of many of these receptors and cellular binding proteins have not been fully explored in RA-responsive human neuroblastoma cells. We have previously shown that RAR transcripts and protein are present in human neuroblastoma cells, and that all-trans RA induces the expression of the RAR beta mRNA. In this paper, we demonstrate that human neuroblastoma cells express mRNA for RXR alpha and beta. The mRNA for CRABPI is present in untreated human neuroblastoma cells, whereas the mRNA for CRABPII is induced in cells treated with either all-trans RA or 9-cis RA. Furthermore, 9-cis RA, a ligand that binds to both the RAR and the RXR families, selectively activates the CRABPII gene. In contrast, all-trans RA and 9-cis RA are equally effective in the induction of RAR beta transcript and inhibition of cell proliferation. Since both retinoids inhibit human neuroblastoma cell proliferation, it appears that induction of RAR beta rather than of CRABPII is more likely linked to the regulation of human neuroblastoma cell growth.


Subject(s)
Neuroblastoma/metabolism , Receptors, Retinoic Acid/biosynthesis , Tretinoin/pharmacology , Base Sequence , Cell Division/drug effects , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Neuroblastoma/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Receptors, Retinoic Acid/genetics , Tumor Cells, Cultured
16.
Dev Dyn ; 205(1): 52-63, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8770551

ABSTRACT

In explanted embryonic chick sympathetic neurons, all-trans retinoic acid (RA) as well as nerve growth factor (NGF) were found to be required for neuronal survival and neurite outgrowth at early stages of development (day 6.5-7) in agreement with previous work (Rodriguez-Tébar and Rohrer [1991] Development 112:813-820). The dependence of neurons on all-trans RA for survival diminished at later stages of development. However, all-trans RA was found to be needed at all stages of development in order to maximize neurite outgrowth. Further, removal of all-trans RA from the cultures led to a rapid degeneration of the formed neurites, demonstrating the essentiality of all-trans RA for both the development of neurites, and for the maintenance of existing neurites in cultured embryonic sympathetic neurons. The mechanism whereby all-trans RA exerts its effects on embryonic sympathetic neurons may involve activation of the nuclear retinoic acid and retinoid-X receptor (RAR and RXR) families. The results of Northern blot analyses and/or reverse transcriptase-polymerase chain reaction (RT-PCR) studies show that embryonic sympathetic ganglia express RAR beta, RAR gamma and RXR gamma mRNAs. RXR gamma mRNA is expressed at highest levels in immature neurons that are not yet responsive to NGF (day 6.5-7) and message levels decline with increasing developmental age. In contrast, RAR beta transcript levels are barely detectable at day 6.5-7, and increase approximately 4-fold in ganglia from embryos at day 8.5-9 and decline thereafter. RT-PCR studies show that RAR gamma mRNA is expressed both early (day 6.5-7) and late (day 15) in ganglionic development. Transcripts for the NGF receptors, p75NGFR and p140trk were also examined. The appearance of a single 2.7 kb p140trk transcript coincides with the time when RAR beta mRNA is maximally expressed, raising the possibility that NGF receptors may be targets of retinoid action. Evidence is also presented that all-trans RA may enhance neurite outgrowth by mechanisms other than simply inducing NGF-responsiveness of neurons.


Subject(s)
Ganglia, Sympathetic/drug effects , Neurites/drug effects , Tretinoin/pharmacology , Animals , Base Sequence , Cells, Cultured , Chick Embryo , DNA Primers/genetics , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/ultrastructure , Molecular Sequence Data , Nerve Growth Factors/pharmacology , Neurites/ultrastructure , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Proc Natl Acad Sci U S A ; 90(15): 7293-7, 1993 Aug 01.
Article in English | MEDLINE | ID: mdl-8394016

ABSTRACT

Competition of all-trans-retinol and all-trans-retinaldehyde with 3H-labeled all-trans-retinoic acid (RA) for binding to retinoic acid receptors (RARs) was examined in human neuroblastoma cell nuclear extracts. All-trans-retinol was 35-fold less potent than all-trans-RA, whereas all-trans-retinaldehyde was 500-fold less active in binding to the nuclear receptors. To confirm that all-trans-retinol binds to RARs, experiments were carried out with RARs alpha, beta, and gamma expressed as bacterial fusion proteins. All-trans-retinol was only 4- to 7-fold less potent than all-trans-RA in binding to all three RAR subtypes. The all-trans-retinol binding observed was not the result of metabolism of retinol to RA or some other active compound during the binding experiment. Retinyl acetate was virtually inactive in competition binding experiments, while very slight activity was observed with 13-cis-RA and all-trans-retinaldehyde. Significant competition occurred with 4-hydroxy-RA and 4-keto-RA, which were 15- to 40-fold less potent than all-trans-RA. The 9-cis isomer of RA was equipotent with all-trans-retinol in these studies. These results suggest that all-trans-retinol cannot be excluded as a physiologically significant ligand for RAR-mediated gene expression.


Subject(s)
Carrier Proteins/metabolism , Vitamin A/metabolism , Base Sequence , Binding, Competitive , Cell Nucleus/metabolism , Humans , In Vitro Techniques , Ligands , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Receptors, Retinoic Acid , Recombinant Fusion Proteins/metabolism , Tretinoin/metabolism , Tumor Cells, Cultured
18.
Teratology ; 62(6): 374-84, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11091359

ABSTRACT

BACKGROUND: Normal embryonic development and survival in utero is dependent on an adequate supply of vitamin A. Embryos from vitamin A-deficient (VAD) pregnant rats fed an inadequate amount of all-trans retinoic acid (atRA; 12 microg per g of diet or approximately 230 microg per rat per day) exhibit severe developmental abnormalities of the anterior cardinal vein and hindbrain by embryonic day (E) 12.5 and die shortly thereafter. METHODS: In the present study, we sought to determine whether supplementation of VAD-RA supported (12 microg per g of diet) pregnant rats with retinol (ROL) at the late-gastrula (presomite or rat E9.5) or early somite stages (E10.5), or provision of higher levels of atRA throughout this period could prevent abnormalities in the developing cardiovascular and nervous systems. RESULTS: A newly described defect in the sinuatrial venus valve along with enlarged anterior cardinal veins and nervous system abnormalities and the later death of embryos are prevented by supplementing pregnant animals with ROL on the morning of E9.5. If ROL supplementation is delayed by 1 day (E10.5), most embryos are abnormal and die by E18.5. Supplementation of VAD rats with atRA (250 microg per g of diet) between E8.5 and E10.5 also prevents the cardiovascular and nervous system abnormalities and a significant number of these embryos survive to parturition. Thus, high levels of atRA can obviate the need for ROL between E9.5 and E10.5. CONCLUSIONS: These results support an essential role for retinoid signaling between the late gastrula and early somite stages in the rat embryo for normal morphogenesis of the primitive heart tube and the posterior hindbrain. Further, these results suggest that embryonic death occurring at midgestation in the VAD rat may be linked to the abnormal development of one or both of these embryonic structures.


Subject(s)
Abnormalities, Multiple/etiology , Fetal Heart/drug effects , Fetal Resorption/etiology , Pregnancy Complications/physiopathology , Rhombencephalon/abnormalities , Tretinoin/therapeutic use , Veins/abnormalities , Vitamin A Deficiency/physiopathology , Vitamin A/analogs & derivatives , Abnormalities, Multiple/prevention & control , Animal Feed , Animals , Cranial Nerves/abnormalities , Cranial Nerves/embryology , Diterpenes , Dose-Response Relationship, Drug , Embryonic and Fetal Development/drug effects , Female , Fetal Death/etiology , Fetal Death/prevention & control , Fetal Resorption/prevention & control , Gastrula/drug effects , Genes, Homeobox , Gestational Age , Morphogenesis/drug effects , Pregnancy , Pregnancy Complications/blood , Rats , Retinyl Esters , Rhombencephalon/embryology , Transcription Factors/genetics , Tretinoin/administration & dosage , Veins/embryology , Vitamin A/administration & dosage , Vitamin A/therapeutic use , Vitamin A Deficiency/blood
19.
Dev Biol ; 220(2): 263-84, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10753515

ABSTRACT

The developing nervous system is particularly vulnerable to vitamin A deficiency. Retinoid has been proposed to be a posteriorizing factor during hindbrain development, although direct evidence in the mammalian embryo is lacking. In the present study, pregnant vitamin A-deficient (VAD) rats were fed purified diets containing varying levels of all-trans-retinoic acid (atRA; 0, 0.5, 1.5, 6, 12, 25, 50, 125, or 250 microg/g diet) or were supplemented with retinol. Hindbrain development was studied from embryonic day 10 to 12.5 ( approximately 6 to 40 somites). Normal morphogenesis was observed in all embryos from groups fed 250 microg atRA/g diet or retinol. The most caudal region of the hindbrain was the most sensitive to retinoid insufficiency, as evidenced by a loss of the hypoglossal nerve (cranial nerve XII) in embryos from the 125 microg atRA/g diet group. Further reduction of atRA to 50 microg/g diet led to the loss of cranial nerves IX, X, XI, and XII and associated sensory ganglia IX and X in all embryos as well as the loss of hindbrain segmentation caudal to the rhombomere (r) 3/4 border in a subset of embryos. Dysmorphic orthotopic otic vesicles or immature otic-like vesicles in both orthotopic and caudally ectopic locations were also observed. As the level of atRA was reduced, a loss of caudal hindbrain segmentation was observed in all embryos and the incidence of otic vesicle abnormalities increased. Perturbations in hindbrain segmentation, cranial nerve formation, and otic vesicle development were associated with abnormal patterning of the posterior hindbrain. Embryos from VAD dams fed between 0.5 and 50 microg atRA/g diet exhibited Hoxb-1 protein expression along the entire neural tube caudal to the r3/r4 border at a time when it should be restricted to r4. Krox-20 protein expression was expanded in r3 but absent or reduced in presumptive r5. Hoxd-4 mRNA expression was absent in the posterior hindbrain, and the rostral limit of Hoxb-5 protein expression in the neural tube was anteriorized, suggesting that the most posterior hindbrain region (r7/r8) had been deleted and/or improperly patterned. Thus, when limiting amounts of atRA are provided to VAD dams, the caudal portion of the hindbrain is shortened and possesses r4/r5-like characteristics, with this region finally exhibiting r4-like gene expression when retinoid is restricted even more severely. Thus, regions of the anterior hindbrain (i.e., r3 and r4) appear to be greatly expanded, whereas the posterior hindbrain (r5-r8) is reduced or absent. This work shows that retinoid plays a critical role in patterning, segmentation, and neurogenesis of the caudal hindbrain and serves as an essential posteriorizing signal for this region of the central nervous system in the mammal.


Subject(s)
Rhombencephalon/embryology , Vitamin A Deficiency/embryology , Animals , Biomarkers , Cranial Nerves/embryology , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Ear/embryology , Early Growth Response Protein 2 , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Models, Biological , Rats , Rats, Sprague-Dawley , Rhombencephalon/abnormalities , Rhombencephalon/drug effects , Time Factors , Transcription Factors/metabolism , Tretinoin/pharmacology , Vitamin A/pharmacology , Vitamin A/physiology
20.
J Nutr ; 129(12): 2246-50, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10573558

ABSTRACT

Interactions between vitamin A and vitamin D have been suggested for several decades but have not been established. In particular, vitamin A has been proposed to intensify the severity of the bone mineralization disease, rickets and inhibit the ability of vitamin D to cure this disease. To investigate this hypothesis, weanling Holtzman rats were fed a 1.2% calcium, 0.1% phosphorus diet and 15.5 ng ergocalciferol (vitamin D(2)) every 3 d for 21 d in the presence of increasing amounts of retinyl acetate (0 microg to 8621 microg/d). The increasing amounts of retinyl acetate produced a progressive and significant decrease in total bone ash (P < 0.001) and an increase in epiphyseal plate width (P < 0.001). The same experiment conducted with increasing amounts of vitamin D(2) (0 to 645 ng/d) indicated that the antagonism by retinyl acetate could be demonstrated at all vitamin D(2) dosages. To further investigate this antagonistic relationship, weanling Holtzman rats were fed a 0. 47% calcium, 0.3% phosphorus diet and 15.5 ng vitamin D(2) every 3 d for 33 d in the presence of increasing retinyl acetate (0 to 3448 microg/d). In the absence of retinyl acetate, these rats maintained a normal serum calcium level (2.34 mmol/L). Increasing retinyl acetate, however, eliminated the ability of vitamin D(2) to elevate the level of serum calcium (1.35 mmol/L). These results illustrated in vivo antagonism of vitamin D(2) action on intestine and bone by retinyl acetate.


Subject(s)
Vitamin A/pharmacology , Vitamin D/antagonists & inhibitors , Animals , Calcium/blood , Diterpenes , Growth Plate/drug effects , Growth Plate/growth & development , Male , Minerals/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Retinyl Esters , Vitamin A/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL