Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Cancer Chemother Pharmacol ; 73(1): 103-11, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24178368

ABSTRACT

PURPOSE: This study characterized the multiple-dose pharmacokinetics of vemurafenib 240-960 mg twice daily (bid) in BRAF(V600E) mutation-positive metastatic melanoma patients, using the commercial formulation (240-mg microprecipitated bulk powder film-coated tablets). METHODS: Melanoma patients (N = 52) were randomly allocated to four vemurafenib dose cohorts (240, 480, 720, or 960 mg bid for 14 days). After the day 15 morning dose, doses were interrupted until day 22, at which point patients were restarted on vemurafenib. Serial pharmacokinetic samples were collected after the morning dose on days 1, 9, and 15; trough pharmacokinetic samples were collected on day 2. RESULTS: Vemurafenib concentration increased with multiple doses to steady state at day 15; C(max), AUC(0-8h), and AUC(0-168h) increased between 3.3- and 3.8-fold across the fourfold dose range tested. Statistical analysis indicated dose proportionality across the dose range of 240-960 mg bid. Day 15 mean accumulation ratios (ratio of AUC(0-8h) on day 15/AUC(0-8h) on day 1) ranged from ~19 to 25 across cohorts. At steady state, the peak-to-trough ratio for vemurafenib exhibited a relatively flat concentration-time profile throughout the bid dosing interval. During dose interruption (days 15-22), mean vemurafenib trough concentrations decreased to minimal levels; vemurafenib exhibited a mean terminal phase half-life of 31.5-38.4 h. CONCLUSIONS: Vemurafenib plasma concentration accumulates with multiple bid doses of 240 mg. Vemurafenib exposure (AUC and C(max)) is dose proportional over the 240- to 960-mg bid dose range and exhibits constant drug levels over the bid dosing interval.


Subject(s)
Indoles/pharmacokinetics , Melanoma/drug therapy , Mutation , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacokinetics , Adult , Aged , Area Under Curve , Female , Humans , Indoles/adverse effects , Male , Melanoma/genetics , Melanoma/secondary , Middle Aged , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/adverse effects , Vemurafenib
2.
Antimicrob Agents Chemother ; 57(12): 6097-105, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24080649

ABSTRACT

Ribavirin (RBV) is an integral part of standard-of-care hepatitis C virus (HCV) treatments and many future regimens under investigation. The pharmacokinetics (PK), safety, and tolerability of RBV in chronically HCV-infected patients with renal impairment are not well defined and were the focus of an open-label PK study in HCV-infected patients receiving RBV plus pegylated interferon. Serial RBV plasma samples were collected over 12 h on day 1 of weeks 1 and 12 from patients with moderate renal impairment (creatinine clearance [CLCR], 30 to 50 ml/min; RBV, 600 mg daily), severe renal impairment (CLCR, <30 ml/min; RBV, 400 mg daily), end-stage renal disease (ESRD) (RBV, 200 mg daily), or normal renal function (CLCR, >80 ml/min; RBV, 800 to 1,200 mg daily). Of the 44 patients, 9 had moderately impaired renal function, 10 had severely impaired renal function, 13 had ESRD, and 12 had normal renal function. The RBV dose was reduced because of adverse events (AEs) in 71% and 53% of severe and moderate renal impairment groups, respectively. Despite this modification, patients with moderate and severe impairment had 12-hour (area under the concentration-time curve from 0 to 12 h [AUC0-12]) values 36% (38,452 ng · h/ml) and 25% (35,101 ng · h/ml) higher, respectively, than those with normal renal function (28,192 ng · h/ml). Patients with ESRD tolerated a 200-mg daily dose, and AUC0-12 was 20% lower (22,629 ng · h/ml) than in patients with normal renal function. PK modeling and simulation (M&S) indicated that doses of 200 mg or 400 mg alternating daily for patients with moderate renal impairment and 200 mg daily for patients with severe renal impairment were the most appropriate dose regimens in these patients.


Subject(s)
Antiviral Agents/pharmacokinetics , Hepatitis C, Chronic/drug therapy , Interferon-alpha/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Renal Insufficiency/drug therapy , Ribavirin/pharmacokinetics , Adult , Aged , Antiviral Agents/blood , Antiviral Agents/pharmacology , Area Under Curve , Drug Administration Schedule , Drug Dosage Calculations , Female , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/blood , Interferon-alpha/pharmacology , Male , Metabolic Clearance Rate , Middle Aged , Polyethylene Glycols/pharmacology , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Renal Insufficiency/blood , Renal Insufficiency/complications , Renal Insufficiency/virology , Ribavirin/blood , Ribavirin/pharmacology , Severity of Illness Index
3.
Aliment Pharmacol Ther ; 35(10): 1209-20, 2012 May.
Article in English | MEDLINE | ID: mdl-22469033

ABSTRACT

BACKGROUND: Amongst Caucasian, Hispanic and African Americans with genotype 1 hepatitis C virus (HCV), there is a wide variation in response to treatment with peginterferon alfa-2a (PEG-IFN alfa-2a) and ribavirin. AIM: To evaluate the pharmacokinetics (PK) of PEG-IFN alfa-2a and ribavirin among these three groups. METHODS: Forty-seven patients with genotype 1 CHC (17 African Americans, 14 Hispanics and 16 Caucasians) received 8 weeks of PEG-IFN alfa-2a (180 µg/week) and ribavirin (1000 or 1200 mg/day). PEG-IFN alfa-2a serum concentrations and ribavirin plasma concentrations were measured following the first dose and at week 8. Pharmacokinetic parameters (C(max), T(max), AUC, CL/F) were estimated using noncompartmental methods. RESULTS: There was no difference in the pharmacokinetic parameters for PEG-IFN alfa-2a following single-dose or steady-state administration between African American or Hispanic patients compared with Caucasian patients. Ribavirin pharmacokinetic parameters were similar between Hispanic and Caucasian patients for single-dose and steady-state administration. The single-dose C(max) was 33% lower (P < 0.05) in African American compared with Caucasian patients. Other ribavirin single-dose and steady-state pharmacokinetic parameters were slightly decreased (approximately 20% lower) in African American patients, but were not considered clinically meaningful. CONCLUSIONS: No differences were observed in PEG-IFN alfa-2a pharmacokinetic parameters between African American or Hispanic patients compared with Caucasian patients. For ribavirin, no differences were observed in pharmacokinetic parameters between Hispanic and Caucasian patients. While a trend towards increased ribavirin clearance and decreased exposure was observed in African American patients vs. Caucasian patients, the differences were small and not considered clinically meaningful (Clinical Trial Number: NP17354).


Subject(s)
Antiviral Agents/pharmacokinetics , Hepatitis C, Chronic/metabolism , Interferon-alpha/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Ribavirin/pharmacokinetics , Adult , Black or African American/genetics , Antiviral Agents/administration & dosage , Area Under Curve , Clinical Trials as Topic , Dose-Response Relationship, Drug , Female , Genotype , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/genetics , Hispanic or Latino/genetics , Humans , Interferon-alpha/administration & dosage , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Ribavirin/administration & dosage , White People/genetics
4.
Clin Cancer Res ; 17(3): 611-9, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21127194

ABSTRACT

PURPOSE: This pediatric phase I study was designed to identify the doses of RG1507, a monoclonal antibody against the Type 1 Insulin-like Growth Factor Receptor (IGF1R), that achieves exposures equivalent to those achieved in adults at recommended doses. EXPERIMENTAL DESIGN: Children with relapsed or refractory solid tumors were treated using the same doses and administration schedules of RG1507 (3 and 9 mg/kg/wk, and 16 mg/kg every 3 weeks [q3W]) as those studied in adults. Detailed pharmacokinetic (PK) sampling was performed after the first dose; selected peak and trough levels were subsequently obtained. Target exposures were ≥85% of mean areas under concentration x time curves (AUCs) in adults at doses of 9 mg/kg/wk and 16 mg/kg q3W. A maximum tolerated dose could be identified if dose-limiting toxicities (DLT) occurred. RESULTS: Thirty-one evaluable patients aged 3-17 years were enrolled at 3 mg/kg/wk (n = 3), 9 mg/kg/wk (n = 18), or 16 mg/kg q3W (n = 10). There were no DLTs. At 9 mg/kg/wk the mean AUC(0-7d) (21,000 µg h/mL) exceeded the target (16,000 µg h/mL). At 16 mg/kg q3W, the mean AUC(021d) (70,000 µg h/mL) exceeded the target (59,400 µg h/mL). Clearance normalized to body weight was age dependent. There were no objective responses. Seven patients had stable disease for >12 weeks, including two patients with osteosarcoma with stable disease for 52+ and 78+ weeks. CONCLUSIONS: The recommended doses of RG1507 in children with solid tumors are 9 mg/kg/wk and 16 mg/kg q3W. This flexible design is well suited for trials of agents associated with limited toxicity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Receptor, IGF Type 1/antagonists & inhibitors , Adolescent , Antibodies, Monoclonal/pharmacokinetics , Child , Child, Preschool , Drug Resistance, Neoplasm , Female , Humans , Male , Recurrence
5.
Diabetes ; 50(3): 622-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11246883

ABSTRACT

Transgenic mice that overexpress the entire glucokinase (GK) gene locus have been previously shown to be mildly hypoglycemic and to have improved tolerance to glucose. To determine whether increased GK might also prevent or diminish diabetes in diet-induced obese animals, we examined the effect of feeding these mice a high-fat high-simple carbohydrate low-fiber diet (HF diet) for 30 weeks. In response to this diet, both normal and transgenic mice became obese and had similar BMIs (5.3 +/- 0.1 and 5.0 +/- 0.1 kg/m2 in transgenic and non-transgenic mice, respectively). The blood glucose concentration of the control mice increased linearly with time and reached 17.0 +/- 1.3 mmol/l at the 30th week. In contrast, the blood glucose of GK transgenic mice rose to only 9.7 +/- 1.2 mmol/l at the 15th week, after which it returned to 7.6 +/- 1.0 mmol/l by the 30th week. The plasma insulin concentration was also lower in the GK transgenic animals (232 +/- 79 pmol/l) than in the controls (595 +/- 77 pmol/l), but there was no difference in plasma glucagon concentrations. Together, these data indicate that increased GK levels dramatically lessen the development of both hyperglycemia and hyperinsulinemia associated with the feeding of an HF diet.


Subject(s)
Chromosome Mapping , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Glucokinase/genetics , Obesity/complications , Transgenes/physiology , Animals , Blood Glucose/analysis , Dietary Fats/administration & dosage , Glucagon/blood , Glucokinase/metabolism , Insulin/blood , Liver/enzymology , Mice , Mice, Transgenic/genetics , Obesity/blood , Obesity/etiology , RNA, Messenger/metabolism , Reference Values
6.
J Biol Chem ; 275(11): 7826-31, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10713097

ABSTRACT

The glucokinase regulatory protein (GKRP) inhibits glucokinase competitively with respect to glucose by forming a protein-protein complex with this enzyme. The physiological role of GKRP in controlling hepatic glucokinase activity was addressed using gene targeting to disrupt GKRP gene expression. Heterozygote and homozygote knockout mice have a substantial decrease in hepatic glucokinase expression and enzymatic activity as measured at saturating glucose concentrations when compared with wild-type mice, with no change in basal blood glucose levels. Interestingly, when assayed under conditions to promote the association between glucokinase and GKRP, liver glucokinase activity in wild-type and null mice displayed comparable glucose phosphorylation capacities at physiological glucose concentrations (5 mM). Thus, despite reduced hepatic glucokinase expression levels in the null mice, glucokinase activity in the liver homogenates was maintained at nearly normal levels due to the absence of the inhibitory effects of GKRP. However, following a glucose tolerance test, the homozygote knockout mice show impaired glucose clearance, indicating that they cannot recruit sufficient glucokinase due to the absence of a nuclear reserve. These data suggest both a regulatory and a stabilizing role for GKRP in maintaining adequate glucokinase in the liver. Furthermore, this study provides evidence for the important role GKRP plays in acutely regulating of hepatic glucose metabolism.


Subject(s)
Carrier Proteins , Glucokinase/antagonists & inhibitors , Glucose/metabolism , Liver/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Age Factors , Animals , Enzyme Inhibitors/metabolism , Gene Targeting , Glucose Tolerance Test , Heterozygote , Homeostasis , Homozygote , Insulin/blood , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Mutagenesis , Proteins/genetics
7.
J Biol Chem ; 274(52): 37125-30, 1999 Dec 24.
Article in English | MEDLINE | ID: mdl-10601273

ABSTRACT

Hepatic glucokinase (GK) moves between the nucleus and cytoplasm in response to metabolic alterations. Here, using heterologous cell systems, we have found that at least two different mechanisms are involved in the intracellular movement of GK. In the absence of the GK regulatory protein (GKRP) GK resides only in the cytoplasm. However, in the presence of GKRP, GK moves to the nucleus and resides there in association with this protein until changes in the metabolic milieu prompt its release. GK does not contain a nuclear localization signal sequence and does not enter the nucleus in a GKRP-independent manner because cells treated with leptomycin B, a specific inhibitor of leucine-rich NES-dependent nuclear export, do not accumulate GK in the nucleus. Instead, entry of GK into the nucleus appears to occur via a piggy-back mechanism that involves binding to GKRP. Nuclear export of GK, which occurs after its release from GKRP, is due to a leucine-rich nuclear export signal within the protein ((300)ELVRLVLLKLV(310)). Thus, GKRP appears to function as both a nuclear chaperone and metabolic sensor and is a critical component of a hepatic GK translocation cycle for regulating the activity of this enzyme in response to metabolic alterations.


Subject(s)
Carrier Proteins , Cell Nucleus/enzymology , Glucokinase/metabolism , Liver/enzymology , Protein Sorting Signals , Proteins/physiology , Adaptor Proteins, Signal Transducing , Biological Transport , Glucokinase/chemistry , HeLa Cells , Humans
8.
Farmaco ; 53(4): 262-5, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9658583

ABSTRACT

The environment for developing novel therapeutic agents has undergone dramatic change over the past decade. Innovative strategies for identifying and utilizing molecular targets linked to particular human diseases are replacing the classic approach of screening chemical compounds for potential therapeutic action on unknown targets. Since genetic components are involved in many known diseases, mouse and human genetics, positional cloning and other molecular biology-based approaches are now used to identify genes that are associated with these diseases. It is thought that identification of these disease-linked genes may lead to the discovery and understanding of the physiologically relevant biochemical pathways underlying the disease processes. Clearly, a knowledge of these biochemical pathways will provide future molecular targets, enzymes or receptors, that will offer opportunities to apply modern methods of high throughput screening, medicinal chemistry, parallel synthesis and combinatorial chemistry for drug discovery. In this manuscript, we illustrate how mouse genetics and molecular biology-based approaches have led to the identification of all five known single gene mutations that cause obesity in mice. Additionally, we describe how identification of these genes has helped unravel underlying biochemical pathways that regulate behavioral, metabolic and neuroendocrine responses in rodents.


Subject(s)
Obesity/genetics , Animals , Energy Metabolism , Gene Targeting , Humans , Mice , Mice, Obese , Mutation , Obesity/drug therapy
9.
Leukemia ; 12(4): 554-62, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9557614

ABSTRACT

The chimeric receptor, RARalpha/VDR, contains the DNA-binding domain of the retinoic acid receptor (RARalpha) and the ligand-binding domain of the vitamin D receptor (VDR). The ligand-binding properties of RARalpha/VDR are equivalent to that of VDR, with an observed Kd for 1alpha,25 dihydroxy-vitamin D3 (D3) of 0.5 nM. In CV-1 cells, both RARalpha and RARalpha/VDR induce comparable levels of ligand-mediated transcriptional activity from the retinoic acid responsive reporter gene, beta(RARE)3-TK-luciferase, in the presence of the ligand predicted from the receptor ligand-binding domain. Two chimeric RAR receptors were constructed which contained the ligand-binding domain of the estrogen receptor (ER): RARalpha/ER and ER/RARalpha/ER. Both RARalpha/ER and ER/RARalpha/ER bind beta-estradiol with high affinity, and are transcriptionally active only from palindromic RAREs (TREpal and/or (TRE3)3). Only RARalpha/VDR matched in kind and degree the functional characteristics of RARalpha: (1) maximally active from the beta(RARE); (2) moderately active from the TREs; (3) inactive from the retinoic X receptor response elements (RXREs) ApoA1 and CRBP II; (4) forms heterodimers with RXRalpha; and (5) binds to the betaRARE. F9 embryonal carcinoma cell lines were generated which express RARalpha/VDR mRNA (F9RARalpha/VDR cells) and compared with F9 wild-type (F9-Wt) cells, which do not express VDR mRNA. Treatment with all-trans retinoic acid (tRA) inhibits cell growth and induces the differentiation morphology in both F9-Wt and F9-RARalpha/VDR cells; whereas, treatment with D3 is similarly effective only for F9-RARalpha/VDR cells. It is concluded RARalpha/VDR is an useful 'tool' to pinpoint, or to augment transcription from RAREs in gene pathways controlled by RAR without inhibiting the retinoid responsiveness of endogenous RARs.


Subject(s)
Receptors, Calcitriol/physiology , Receptors, Retinoic Acid/physiology , Recombinant Fusion Proteins/physiology , Animals , COS Cells , Cell Differentiation/physiology , Cholecalciferol/metabolism , Dimerization , Estradiol/metabolism , Kinetics , Mice , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Estrogen/physiology , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors , Substrate Specificity , Transcription Factors/metabolism , Transcriptional Activation/physiology , Transfection
10.
FASEB J ; 10(13): 1524-31, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8940298

ABSTRACT

We previously identified a carboxy-terminal transactivation function termed AF-2 within the last 15 amino acids of the ligand binding domain of the human retinoic acid receptor alpha (hRAR alpha). Truncation of this region abolished transcriptional activity. Here we provide a systematic analysis using alanine scanning mutagenesis of amino acids from Ser405 to Gly419 on a truncated hRAR alpha (delta419) to identify residues within this region that are responsible for transcriptional activity. Whereas mutations in positions 405, 408, 411, and 415-419 have little or no effect on the ability of modified receptors to activate a DR5 response element, mutations in positions 406, 407, 409, 410, and 412-413 modify either the potency or efficacy of all-trans retinoic acid (tRA) -induced gene transcription. Therefore, receptors with mutations in positions 409, 410, 413, and 414 have low transcriptional activity over a wide range of tRA concentrations. Receptors with mutations in positions 406, 407, and 412 exhibit a maximum transcriptional activity similar to wild-type hRAR alpha, but require higher concentrations of tRA. Replacing residues 405-419 on delta419 with the conserved AF-2 domain from the vitamin D3 receptor or the estrogen receptor results in a receptor with wild-type or low transcriptional activity, respectively. A full-length hRAR alpha mutant with an alanine substitution at position 406 (hRAR alpha M406A) binds tRA, but unlike the truncated M406A, which lacks the "F" region, it is not transcriptionally active. Protease mapping experiments detect a consistent difference in the conformation of hRAR alpha M406A compared to wild-type hRAR alpha. These data define amino acids from Ser405 to Gly419 on delta419 that are critical for transcriptional activity and point to the importance of the conformational integrity of receptor domains in maintaining ligand-induced transcriptional activation.


Subject(s)
Protein Conformation , Receptors, Retinoic Acid/metabolism , Transcriptional Activation , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Endopeptidases/metabolism , Humans , Mutagenesis , Peptide Mapping , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoic Acid Receptor alpha , Tumor Cells, Cultured
11.
J Biol Chem ; 271(34): 20346-52, 1996 Aug 23.
Article in English | MEDLINE | ID: mdl-8702769

ABSTRACT

The growth of estrogen receptor (ER)-positive breast cancer cells is inhibited by all-trans-retinoic acid (RA). In the present study, estrogen (E2) induction of pS2 mRNA levels was significantly reduced within 6 h following cotreatment with RA. In transient transfection experiments, RA repressed transactivation from a vitellogenin E2-responsive element by approximately 50% and wild-type RA receptor alpha (RARalpha) or RARbeta enhanced this inhibition. Transfection of truncated RARalpha mutants terminating before or at amino acid 412 markedly decreased RA inhibition of E2-induced reporter gene activity. Expression of RARs with deletions of amino acids 413 and 414 in the transactivation-2 (AF-2) domain also reduced RA inhibition, while deletions and point mutations beyond amino acid 414 behaved like the wild-type RARalpha. RA-treated MCF-7 cells transfected with an RARalpha AF-2 region mutant were twice as sensitive to growth inhibition as untransfected and vector-transfected control cells. Thus, the AF-2 domain in the C terminus of the RARalpha mediates RA inhibition of ER-induced transcription in breast cancer cells. In addition, transcriptional interference between RARs and ERs may contribute to RA inhibition of ER-positive breast cancer cell growth.


Subject(s)
Neoplasm Proteins/genetics , Proteins , Receptors, Estrogen/antagonists & inhibitors , Receptors, Retinoic Acid/chemistry , Tretinoin/pharmacology , Base Sequence , DNA Primers/chemistry , DNA-Binding Proteins/metabolism , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , RNA, Messenger/genetics , Receptors, Retinoic Acid/physiology , Regulatory Sequences, Nucleic Acid , Retinoic Acid Receptor alpha , Structure-Activity Relationship , Transcription, Genetic , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
12.
Cell Growth Differ ; 7(3): 327-37, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8838863

ABSTRACT

Retinoic acid (RA) is a potent inducer of differentiation of embryonal carcinoma PCC4.aza1R cells into mesenchymal stem cells. Induction of Hoxa-1, Hoxa-5, cellular retinoic acid-binding protein (CRABP) I and II, and retinoic acid receptor (RAR)-beta expression occurs early in this multistage program of differentiation. RA is also a potent inducer of these genes in the differentiation-defective mutant PCC4(RA)-1; however, RA is much less effective in the mutant cell line PCC4(RA)-2. The up-regulation of several of these genes by RA is, at least in part, due to increased transcription. It is likely that some of these changes are mediated either directly or indirectly by nuclear retinoid receptors. Previously, we characterized the expression of RARs in PCC4.aza1R and (RA)-1 and (RA)-2 cells. In this study, we show that these cells also express retinoid X receptor (RXR)-alpha, RXR-beta, and RXR-gamma and that RA treatment down-regulates the expression of RXR-gamma. No large differences were found in RXR mRNA expression between parental and mutant cell lines except that PCC4(RA)-1 cells expressed an 8-fold higher level of RXR gamma mRNA than the parental cells. To obtain more insight into the retinoid signaling pathways involved in the regulation of this pathway of differentiation, we examined the action of two retinoid receptor-selective agonists and one antagonist. The RAR-selective retinoid SRI-6751-84 is a very effective inducer of transactivation of beta RARE-tk-LUC, but not of RXRE-tk-CAT, in PCC4.aza1R cells and is a very potent inducer of morphological differentiation and Hoxa-1, Hoxa-5, CRABP II, and RAR-beta expression. In contrast, the RXR-selective retinoid SR11,217, which transactivates the RXRE-tk-CAT effectively, but beta RARE-tk-LUC poorly, is unable to induce differentiation and has little effect on the expression of these early genes. The RAR-alpha-selective antagonist Ro 41-5253, which inhibits RARE-dependent transcriptional activation, has by itself no effect on the differentiation of PCC4.aza1R cells. However, this antagonist is able to block the induction of morphological differentiation by the RAR-selective retinoid as well as the expression of Hoxa-1, Hoxa-5, CRABP II, and RAR-beta. Our data suggest that the activation of RAR signaling pathways is important in initiating the cascade of changes in gene expression that result in the differentiation of PCC4.aza1R into mesenchymal stem cells. In addition, we demonstrate that the two mutant cell lines, PCC4(RA)-1 and PCC4(RA)-2, are defective at different stages of the differentiation program.


Subject(s)
Gene Expression Regulation/drug effects , Neoplastic Stem Cells/cytology , Phosphoproteins , Receptors, Retinoic Acid/genetics , Retinoids/pharmacology , Animals , Benzoates/pharmacology , Cell Differentiation/drug effects , Chromans/pharmacology , Embryonal Carcinoma Stem Cells , Genes, Reporter/genetics , Homeodomain Proteins/genetics , Ligands , Luciferases/genetics , Mice , Neoplastic Stem Cells/drug effects , RNA, Messenger/biosynthesis , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Retinoids/antagonists & inhibitors , Signal Transduction/physiology , Transcription Factors/genetics , Transcriptional Activation , Tretinoin/pharmacology
13.
Mol Cell Biol ; 15(10): 5226-34, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7565671

ABSTRACT

Retinoid X receptors (RXRs), along with retinoic acid (RA) receptors (RARs), mediate the effects of RA on gene expression. Three subtypes of RXRs (alpha, beta, and gamma) which bind to and are activated by the 9-cis stereoisomer of RA have been characterized. They activate gene transcription by binding to specific sites on DNA as homodimers or as heterodimers with RARs and other related nuclear receptors, including the vitamin D receptor, thyroid hormone receptors (TRs), and peroxisome proliferator-activated receptors. Two additional RXR subtypes (delta and epsilon) isolated from zebra fish cDNA libraries are described here; although both subtypes form DNA-binding heterodimers with RARs and TR, neither binds 9-cis RA, and both are transcriptionally inactive on RXR response elements. In cotransfection studies with TR, the delta subtype was found to function in a dominant negative manner, while the epsilon subtype had a slight stimulatory effect on thyroid hormone (T3)-dependent transcriptional activity. The discovery of these two novel receptors in zebra fish expands the functional repertoire of RXRs to include ligand-independent and dominant negative modulation of type II receptor function.


Subject(s)
Gene Expression Regulation, Developmental , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Transcriptional Activation/physiology , Tretinoin/metabolism , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA/metabolism , Genes/genetics , Kinetics , Ligands , Molecular Sequence Data , RNA, Messenger/biosynthesis , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Receptors, Thyroid Hormone/metabolism , Retinoid X Receptors , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/physiology , Triiodothyronine/physiology , Zebrafish/embryology
14.
J Biol Chem ; 270(35): 20258-63, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7657595

ABSTRACT

We have recently identified a small region (amino acids 405-419) within the ligand binding domain of a truncated human retinoic acid receptor alpha (delta 419) that is required for binding of 9-cis-retinoic acid (RA), but not all-trans-retinoic acid (t-RA). To probe the structural determinants of this high affinity 9-cis-RA binding site, a series of delta 419 mutants were prepared whereby an individual alanine residue was substituted for each amino acid within this region. These modified receptors were expressed in mammalian COS-1 cells and assayed for their ability to bind 9-cis-RA as well as t-RA. Only two of the mutants, M406A (mutation of methionine 406 to alanine), and I410A (mutation of isoleucine 410 to alanine) exhibit no detectable binding of 9-cis-RA when analyzed using saturation binding kinetics. Substitution of methionine 406 with the amino acids leucine, isoleucine, and valine yields mutant receptors that exhibit decreased binding for 9-cis-RA as the length or hydrophobicity of the R group decreases. Further substitution of methionine 406 with the small polar amino acid, threonine, results in a loss of detectable 9-cis-RA binding. Since amino acids 405-419 on a human RAR alpha (hRAR alpha) are predicted to form a short amphipathic alpha-helix, modeling of this structure into a helical wheel indicates that these two amino acids, methionine 406 and isoleucine 410, are actually positioned proximal to each other. Data presented here suggest that high affinity 9-cis-RA binding to a hRAR alpha depends on an interaction with the two amino acids methionine 406 and isoleucine 410.


Subject(s)
Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Humans , Kinetics , Molecular Sequence Data , Protein Structure, Secondary , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Restriction Mapping , Retinoic Acid Receptor alpha , Stereoisomerism , Substrate Specificity , Transfection
15.
Leukemia ; 8(11): 1797-806, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7967725

ABSTRACT

The retinoid receptors belong to a large superfamily of ligand-inducible transcription factors that include the steroid, vitamin D and thyroid hormone receptors, the peroxisome proliferator-activated receptor, the insect edysteroid receptor, and a number of orphan receptors whose ligands are unknown. All nuclear receptors have several well-characterized structural domains, including a conserved DNA-binding domain, and a ligand binding domain at the carboxyl terminus of the receptor. The RAR and RXR classes of nuclear retinoic acid receptors are each composed of alpha, beta and gamma subtypes with more than one isoform for each receptor subtype. Data from many investigators suggest there are RAR- and RXR-dependent gene pathways, and that the individual receptor subtypes may control distinct gene expression patterns. In addition, RXR has been found to heterodimerize with other nuclear receptors to form active transcriptional complexes, which influence the activity of a variety of gene pathways important in growth and differentiation. As a result, retinoids have been useful clinical agents in Dermatology and Oncology. However, upon prolonged exposure to retinoic acid, resistance to retinoids has often been encountered both in the clinical setting and in long-term cell culture (HL60R and RAC65 cells). In the latter case, retinoid resistance has been associated with a mutation in the RAR gene which transcribes a RAR receptor truncated at the C-terminal end. These mutated RAR receptors exhibit a reduced affinity for retinoic acid while retaining the ability to bind to a retinoic acid response element on DNA. As a result, these mutant receptors exhibit dominant-negative activity by binding to the DNA without activating transcription and by competing with other receptors for sites on the response element. In fact, dominant-negative activity may be very important in the development of many neoplastic diseases, including acute promyelocytic leukemia (APL), where a t(15;17) chromosomal translocation fuses the PML gene to the RAR gene, to produce a PML-RAR fusion protein in large excess in the cell. However, retinoid resistance in the patient is most probably the result of pharmacokinetic problems, whereby, with continuous retinoid treatment, the plasma levels of retinoic acid gradually decrease to below that required to maintain differentiation of leukemic cells in vivo. A major challenge for drug discovery is to design a drug which circumvents these pharmacokinetic problems either by designing novel drug delivery systems or by employing retinoids which do not bind to CRABP, such as 9-c-RA.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Receptors, Cytoplasmic and Nuclear , Receptors, Retinoic Acid , Retinoids/therapeutic use , Transcription Factors , Animals , DNA-Binding Proteins , Gene Expression Regulation , Humans , Nuclear Proteins , Retinoid X Receptors , Signal Transduction
16.
Development ; 120(9): 2431-42, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7956823

ABSTRACT

Homeobox genes are expressed with a specific spatial and temporal order, which is essential for pattern formation during the early development of both invertebrates and vertebrates. Here we show that widespread ectopic expression of the Hoxa-1 (Hox 1.6) gene directed by a human beta-actin promoter in transgenic mice is embryolethal and produces abnormal phenotypes in a subset of domains primarily located in anterior regions. Interestingly, this abnormal development in the Hoxa-1 transgenic mice is associated with ectopic expression of the Hoxb-1 (Hox 2.9) gene in select hindbrain regions. At gestation day 9.5, two domains of strong Hoxb-1 expression are found in the anterior region of the hindbrains of Hoxa-1 transgenic embryos. One region represents the normal pattern of Hoxb-1 expression in rhombomere 4 and its associated migrating neural crest cells, while another major domain of Hoxb-1 expression consistently appears in rhombomere 2. Similar ectopic domains of beta-galactosidase activity are detected in dual transgenic embryos containing both beta-actin/Hoxa-1 transgene and a Hoxb-1/lacZ reporter construct. Expression of another lacZ reporter gene that directs beta-galactosidase activity predominantly in rhombomere 2 is suppressed in the Hoxa-1 transgenic embryos. We have also detected weaker and variable ectopic Hoxb-1 expression in rhombomeres 1, 3 and 6. No ectopic Hoxb-1 expression is detected in rhombomere 5 and the expression of Hoxa-3 and Krox-20 in this region is unchanged in the Hoxa-1 transgenic embryos. While no obvious change in the morphology of the trigeminal or facial-acoustic ganglia is evident, phenotypic changes do occur in neurons that emanate from rhombomeres 2 and 3 in the Hoxa-1 transgenic embryos. Additionally, alterations in the pattern of Hoxa-2 and Hoxb-1 expression in a subpopulation of neural crest cells migrating from the rhombomere 2 region are detected in these transgenics. Taken together, these data suggest that ectopic Hoxa-1 expression can reorganize select regions of the developing hindbrain by inducing partial transformations of several rhombomeres into a rhombomere-4-like identity.


Subject(s)
Gene Expression Regulation , Genes, Homeobox , Rhombencephalon/embryology , Transformation, Genetic , Actins/genetics , Animals , Base Sequence , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Molecular Sequence Data , Morphogenesis/genetics , Nervous System/embryology , Neural Crest/cytology , Promoter Regions, Genetic
17.
J Biol Chem ; 269(28): 18662-7, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-8034615

ABSTRACT

In order to study the structural details of ligand protein interactions of the human retinoid X receptor alpha (hRXR alpha), the DEF and EF domains of the receptor were expressed as glutathione S-transferase (GST) fusion proteins in Escherichia coli. The fusion proteins were expressed at high levels and were affinity-purified by chromatography over glutathione-agarose. The DEF and EF domains were cleaved from the fusion proteins by digestion with thrombin. Retinoic acid binding was quantitated using two different methods. The apparent dissociation constant (Kd) and the stoichiometry of 9-cis-retinoic acid binding were performed by monitoring quenching of protein fluorescence. To directly compare the binding affinity of the E. coli-derived truncated hRXR alpha with full-length hRXR alpha expressed in transiently transfected COS cells, Scatchard analyses of [3H]9-cis-retinoic acid binding assays were performed. Both methods of analysis indicate that while the cleaved DEF peptide bound 9-cis-retinoic acid tightly, the cleaved EF peptide exhibited variable binding activity between preparations. By fluorimetric analysis, the Kd of the cleaved DEF peptide was estimated to be 3 +/- 0.5 nM with a stoichiometry of 1:1.1 +/- 0.1. By Scatchard analysis, the Kd values for [3H]9-cis-retinoic acid to the GST-hRXR alpha (DEF) peptide and the cleaved DEF peptide were estimated to be 1.8 nM and 5.6 nM, respectively. The estimated molecular mass from high speed sedimentation equilibrium experiments was 36 +/- 2 kDa for the apo-DEF peptide alone and 38 +/- 3 kDa for the holo-DEF peptide complexed with 9-cis-retinoic acid. This suggests that the recombinant ligand binding domain was predominantly in the monomer form. However, dimers of the cleaved DEF peptides were detected in chemical cross-linking experiments both in the presence and absence of 9-cis-retinoic acid. Since the purified E. coli-derived truncated hRXR alpha DEF peptide appears to fully retain its ligand binding activity, it should provide a useful model system for further structural analysis of ligand-protein interactions.


Subject(s)
Escherichia coli/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid , Transcription Factors , Tretinoin/metabolism , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Chromatography, Affinity , Cloning, Molecular/methods , Glutathione Transferase/biosynthesis , Glutathione Transferase/isolation & purification , Glutathione Transferase/metabolism , Humans , Kinetics , Molecular Sequence Data , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Retinoid X Receptors , Spectrometry, Fluorescence , Transfection
18.
Trends Endocrinol Metab ; 5(5): 189-94, 1994 Jul.
Article in English | MEDLINE | ID: mdl-18407207

ABSTRACT

Two classes of nuclear receptors, the retinoic acid receptors (RARs) and the retinoid-X receptors (RXRs), mediate the physiologic activity of retinoids. The RXRs can form biologically active heterodimers with the RARs and with other nuclear receptors, including the vitamin-D, thyroid hormone, and peroxisome proliferator-activated receptors. Thus, the RXRs may play a pivotal role in modulating the action of other hormones or ligands. The RXRs were originally classified as orphan receptors whose cognate ligand was unknown until recently, when 9-cis retinoic acid (9-cis RA) was discovered to bind directly and activate this family of receptors. Since 9-cis RA also binds and activates the RARs, it is interesting to speculate that this natural ligand may regulate a broad range of physiologic processes by mediating transcriptional activity through both RAR- and RXR-linked pathways.

19.
J Biol Chem ; 269(24): 16689-95, 1994 Jun 17.
Article in English | MEDLINE | ID: mdl-8206989

ABSTRACT

Both 9-cis-retinoic acid (RA) and all-trans-RA (t-RA) compete for [3H]9-cis-RA binding to RA receptors (RAR alpha, beta, and gamma) in nucleosol fractions from transiently transfected COS-1 cells with IC50 values of approximately 12 and 5 nM, respectively. Curiously, 9-cis-RA competes for [3H]t-RA binding to mouse RAR alpha, beta, and gamma with IC50 values of 31, 8, and 60 nM, respectively, while t-RA itself does not exhibit such differential competition (IC50 values for RARs, 5 nM). A similar pattern is observed with human retinoic acid receptors (RARs). Differential binding of 9-cis-RA to the RAR beta and gamma receptors is also found following in vitro transcription and translation of these receptors. Displacement assays demonstrate that t-RA exhibits similar off-rates for RAR alpha, beta, and gamma. However, 9-cis-RA is 6-fold more rapidly displaced from RAR gamma than from RAR beta. When RAR-transfected COS-1 cells are incubated with [3H]t-RA, [3H]-9-cis-RA or various mixtures of these two radioligands, high performance liquid chromatography analysis demonstrates that the ligands bound in nucleosol fractions from RAR beta-transfected cells reflect the isomer content of the media. However, in identical whole cell assays, nucleosol fractions from RAR gamma-transfected cells preferentially bind t-RA over 9-cis-RA, consistent with the in vitro data. These binding kinetics in vitro and in whole cells suggest that there could be differences in the interactions of the receptor subtypes with the endogenous retinoic acids under physiologic conditions.


Subject(s)
Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Animals , Binding, Competitive , Cell Line , Chlorocebus aethiops , Kidney , Kinetics , Mice , Protein Biosynthesis , Receptors, Retinoic Acid/biosynthesis , Substrate Specificity , Transcription, Genetic , Transfection , Tritium
20.
Mol Cell Biol ; 14(4): 2323-30, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8139538

ABSTRACT

Retinoids exert their physiological action by interacting with two families of nuclear receptors, the retinoic acid receptors (RARs) and the retinoid X receptors (RXRs), which regulate gene expression by forming transcriptionally active heterodimeric RAR/RXR or homodimeric RXR/RXR complexes on DNA. Retinoid receptor activity resides in several regions, including the DNA and ligand binding domains, a dimerization interface, and both a ligand-independent (AF-1) and a ligand-dependent (AF-2) transactivation function. While 9-cis retinoic acid (RA) alone is the cognate ligand for the RXRs, both 9-cis RA and all-trans RA (t-RA) compete for binding with high affinity to the RARs. This latter observation suggested to us that the two isomers may interact with a common binding site. Here we report that RAR alpha has two distinct but overlapping binding sites for 9-cis RA and t-RA. Truncation of a human RAR alpha to 419 amino acids yields a receptor that binds both t-RA and 9-cis RA with high affinity, but truncation to amino acid 404 yields a mutant receptor that binds only t-RA with high affinity. Remarkably, this region also defines a C-terminal boundary for AF-2, as addition of amino acids 405 to 419 restores receptor-mediated gene activity to a truncated human RAR alpha lacking this region. It is interesting to speculate that binding of retinoid stereoisomers to unique sites within an RAR may function with AF-2 to cause differential activation of retinoid-responsive gene pathways.


Subject(s)
Receptors, Retinoic Acid/metabolism , Transcription Factors , Tretinoin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Cloning, Molecular , Consensus Sequence , DNA, Complementary/metabolism , Gene Expression Regulation , Humans , Macromolecular Substances , Mice , Molecular Sequence Data , Mutagenesis , Oligodeoxyribonucleotides , Protein Conformation , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Retinoid X Receptors , Sequence Deletion , Sequence Homology, Amino Acid , Transcription, Genetic , Transfection , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...