Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 20223, 2023 11 18.
Article in English | MEDLINE | ID: mdl-37980453

ABSTRACT

Several alterations in fibroblast growth factor receptor (FGFR) genes have been found in breast cancer; however, they have not been well characterized as therapeutic targets. Futibatinib (TAS-120; Taiho) is a novel, selective, pan-FGFR inhibitor that inhibits FGFR1-4 at nanomolar concentrations. We sought to determine futibatinib's efficacy in breast cancer models. Nine breast cancer patient-derived xenografts (PDXs) with various FGFR1-4 alterations and expression levels were treated with futibatinib. Antitumor efficacy was evaluated by change in tumor volume and time to tumor doubling. Alterations indicating sensitization to futibatinib in vivo were further characterized in vitro. FGFR gene expression between patient tumors and matching PDXs was significantly correlated; however, overall PDXs had higher FGFR3-4 expression. Futibatinib inhibited tumor growth in 3 of 9 PDXs, with tumor stabilization in an FGFR2-amplified model and prolonged regression (> 110 days) in an FGFR2 Y375C mutant/amplified model. FGFR2 overexpression and, to a greater extent, FGFR2 Y375C expression in MCF10A cells enhanced cell growth and sensitivity to futibatinib. Per institutional and public databases, FGFR2 mutations and amplifications had a population frequency of 1.1%-2.6% and 1.5%-2.5%, respectively, in breast cancer patients. FGFR2 alterations in breast cancer may represent infrequent but highly promising targets for futibatinib.


Subject(s)
Breast Neoplasms , Animals , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Pyrazoles , Pyrimidines/pharmacology , Pyrroles , Receptor, Fibroblast Growth Factor, Type 1/genetics , Disease Models, Animal
2.
Cancers (Basel) ; 13(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638434

ABSTRACT

BRAF-activating mutations are the most frequent driver mutations in papillary thyroid cancer (PTC). Targeted inhibitors such as dabrafenib have been used in advanced BRAF-mutated PTC; however, acquired resistance to the drug is common and little is known about other effectors that may play integral roles in this resistance. In addition, the induction of PTC dedifferentiation into highly aggressive KRAS-driven anaplastic thyroid cancer (ATC) has been reported. We detected a novel RAC1 (P34R) mutation acquired during dabrafenib treatment in a progressive metastatic lesion with ATC phenotype. To identify a potential functional link between this novel mutation and tumor dedifferentiation, we developed a cell line derived from the metastatic lesion and compared its behavior to isogenic cell lines and primary tumor samples. Our data demonstrated that RAC1 mutations induce changes in cell morphology, reorganization of F-actin almost exclusively at the cell cortex, and changes in cell adhesion properties. We also established that RAC1 amplification, with or without mutation, is sufficient to drive cell proliferation and resistance to BRAF inhibition. Further, we identified polyploidy of chromosome 7, which harbors RAC1, in both the metastatic lesion and its derived cell line. Copy number amplification and overexpression of other genes located on this chromosome, such as TWIST1, EGFR, and MET were also detected, which might also lead to dabrafenib resistance. Our study suggests that polyploidy leading to increased expression of specific genes, particularly those located on chromosome 7, should be considered when analyzing aggressive thyroid tumor samples and in further treatments.

3.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507989

ABSTRACT

The phosphoinositide 3-kinase regulatory subunit p85α is a key regulator of kinase signaling and is frequently mutated in cancers. In the present study, we showed that in addition to weakening the inhibitory interaction between p85α and p110α, a group of driver mutations in the p85α N-terminal SH2 domain activated EGFR, HER2, HER3, c-Met, and IGF-1R in a p110α-independent manner. Cancer cells expressing these mutations exhibited the activation of p110α and the AKT pathway. Interestingly, the activation of EGFR, HER2, and c-Met was attributed to the ability of driver mutations to inhibit HER3 ubiquitination and degradation. The resulting increase in HER3 protein levels promoted its heterodimerization with EGFR, HER2, and c-Met, as well as the allosteric activation of these dimerized partners; however, HER3 silencing abolished this transactivation. Accordingly, inhibitors of either AKT or the HER family reduced the oncogenicity of driver mutations. The combination of these inhibitors resulted in marked synergy. Taken together, our findings provide mechanistic insights and suggest therapeutic strategies targeting a class of recurrent p85α mutations.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Catalytic Domain/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/physiology , HCT116 Cells , Humans , Mutation , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Domains/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , src Homology Domains
4.
J Thorac Oncol ; 15(10): 1611-1623, 2020 10.
Article in English | MEDLINE | ID: mdl-32540409

ABSTRACT

INTRODUCTION: Approximately 4% of NSCLC harbor BRAF mutations, and approximately 50% of these are non-V600 mutations. Treatment of tumors harboring non-V600 mutations is challenging because of functional heterogeneity and lack of knowledge regarding their clinical significance and response to targeted agents. METHODS: We conducted an integrative analysis of BRAF non-V600 mutations using genomic profiles of BRAF-mutant NSCLC from the Guardant360 database. BRAF mutations were categorized by clonality and class (1 and 2: RAS-independent; 3: RAS-dependent). Cell viability assays were performed in Ba/F3 models. Drug screens were performed in NSCLC cell lines. RESULTS: A total of 305 unique BRAF mutations were identified. Missense mutations were most common (276, 90%), and 45% were variants of unknown significance. F468S and N581Y were identified as novel activating mutations. Class 1 to 3 mutations had higher clonality than mutations of unknown class (p < 0.01). Three patients were treated with MEK with or without BRAF inhibitors. Patients harboring G469V and D594G mutations did not respond, whereas a patient with the L597R mutation had a durable response. Trametinib with or without dabrafenib, LXH254, and lifirafenib had more potent inhibition of BRAF non-V600-mutant NSCLC cell lines than other MEK, BRAF, and ERK inhibitors, comparable with the inhibition of BRAF V600E cell line. CONCLUSIONS: In BRAF-mutant NSCLC, clonality is higher in known functional mutations and may allow identification of variants of unknown significance that are more likely to be oncogenic drivers. Our data indicate that certain non-V600 mutations are responsive to MEK and BRAF inhibitors. This integration of genomic profiling and drug sensitivity may guide the treatment for BRAF-mutant NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics
5.
Nat Commun ; 9(1): 4583, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389923

ABSTRACT

Activation of platelet-derived growth factor receptor alpha (PDGFRA) by genomic aberrations contributes to tumor progression in several tumor types. In this study, we characterize 16 novel PDGFRA mutations identified from different tumor types and identify three previously uncharacterized activating mutations that promote cell survival and proliferation. PDGFRA Y288C, an extracellular domain mutation, is primarily high mannose glycosylated consistent with trapping in the endoplasmic reticulum (ER). Strikingly, PDGFRA Y288C is constitutively dimerized and phosphorylated in the absence of ligand suggesting that trapping in the ER or aberrant glycosylation is sufficient for receptor activation. Importantly, PDGFRA Y288C induces constitutive phosphorylation of Akt, ERK1/2, and STAT3. PDGFRA Y288C is resistant to PDGFR inhibitors but sensitive to PI3K/mTOR and MEK inhibitors consistent with pathway activation results. Our findings further highlight the importance of characterizing functional consequences of individual mutations for precision medicine.


Subject(s)
Drug Resistance, Neoplasm/genetics , Extracellular Space/chemistry , Molecular Targeted Therapy , Mutation/genetics , Receptor, Platelet-Derived Growth Factor alpha/chemistry , Receptor, Platelet-Derived Growth Factor alpha/genetics , Animals , Cell Line , Cell Proliferation , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/metabolism , Humans , Mice , Phenotype , Protein Domains , Protein Kinase Inhibitors/pharmacology , Signal Transduction
6.
Cancer Cell ; 26(4): 479-94, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25284480

ABSTRACT

PIK3R1 (p85α regulatory subunit of PI3K) is frequently mutated across cancer lineages. Herein, we demonstrate that the most common recurrent PIK3R1 mutation PIK3R1(R348∗) and a nearby mutation PIK3R1(L370fs), in contrast to wild-type and mutations in other regions of PIK3R1, confers an unexpected sensitivity to MEK and JNK inhibitors in vitro and in vivo. Consistent with the response to inhibitors, PIK3R1(R348∗) and PIK3R1(L370fs) unexpectedly increase JNK and ERK phosphorylation. Surprisingly, p85α R348(∗) and L370fs localize to the nucleus where the mutants provide a scaffold for multiple JNK pathway components facilitating nuclear JNK pathway activation. Our findings uncover an unexpected neomorphic role for PIK3R1(R348∗) and neighboring truncation mutations in cellular signaling, providing a rationale for therapeutic targeting of these mutant tumors.


Subject(s)
MAP Kinase Signaling System/drug effects , Mutation , Phosphatidylinositol 3-Kinases/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Nucleus/metabolism , Class Ia Phosphatidylinositol 3-Kinase , Enzyme Activation , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Transport
7.
Invest New Drugs ; 31(1): 30-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22565394

ABSTRACT

This study evaluated the preclinical activity of selumetinib (AZD6244, ARRY-142866), an inhibitor of the mitogen-activated protein kinase kinase (MAPKK or MEK1/2) in 6 nasopharyngeal cancer (NPC) cell lines. Selumetinib could achieve up to 90 % inhibition of cell growth with the respective IC(50) values in NPC cell lines as follow: HK1 = 0.04 µM, HK1-LMP1(B95.8) = 0.17 µM, HONE-1-EBV = 0.46 µM, HONE-1 = 1.79 µM, CNE-2 = 2.20 µM and C666-1 > 10 µM. The drug-sensitive cell lines HK1, HK1-LMP1(B95.8) and HONE-1-EBV have higher basal expression of phosphorylated (pi)-MAPK than the less sensitive cell lines. BRAF mutations were not detected in all 6 cell lines. Re-introduction of the EBV genome into HONE-1 cells, generating the HONE-1-EBV cell line, seemed to result in elevated expression of pi-MAPK and sensitivity to selumetinib when compared with the parental HONE-1 cells. At a concentration of 0.5 µM and 5 µM, selumetinib induced apoptosis (as indicated by cleaved PARP expression and caspase 3 induction), and G(0)/G(1) cycle arrest in HONE-1-EBV and HK1-LMP1(B95.8) cells. The combination of selumetinib (at IC(25) concentration) and the EGFR tyrosine kinase inhibitor, gefitinib (at concentrations of 0.1, 3 and 9 µM) resulted in synergistic growth inhibition in HK1-LMP1(B95.8) cells. The combination of selumetinib (at IC(25) concentration) and cisplatin (at concentrations of 0.1, 0.4, 0.8 and 2 µM) resulted in synergistic growth inhibition in HONE-1 and HONE-1-EBV cells. This result suggests that selumetinib alone or in combination with gefitinib or cisplatin maybe a promising strategy against NPC. Further studies are warranted.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzimidazoles/administration & dosage , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nasopharyngeal Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Synergism , Gefitinib , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Quinazolines/pharmacology
8.
J Orthop Res ; 29(3): 403-13, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20886653

ABSTRACT

Giant cell tumor (GCT) is the most common nonmalignant primary bone tumor reported in Hong Kong. It usually affects young adults between the ages of 20 and 40. This tumor is well known for its potential to recur following treatment. To date no effective adjuvant therapy exists for GCT. Our project aimed to study the effects of pamidronate (PAM), farnesyl transferase inhibitor (FTI-277), geranylgeranyl transferase inhibitor (GGTI-298), and their combinations on GCT stromal cells (SC). Individual treatment with PAM, FTI-277, and GGTI-298, inhibited the cell viability and proliferation of GCT SC in a dose-dependent way. Combination of FTI-277 with GGTI-298 caused synergistic effects in reducing cell viability, and its combination index was 0.49, indicating a strong synergism. Moreover, the combination of FTI-277 with GGTI-298 synergistically enhanced cell apoptosis and activated caspase-3/7, -8, and -9 activities. PAM induced cell-cycle arrest at the S-phase. The combination of PAM with GGTI-298 significantly increased OPG/RANKL mRNA ratio and activated caspase-3/7 activity. Our findings support that the combination of bisphosphonates with GGTIs or FTIs with GGTIs may be used as potential adjuvants in the treatment of GCT of bone.


Subject(s)
Bone Neoplasms , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Giant Cell Tumor of Bone , Osteoprotegerin/genetics , RANK Ligand/genetics , Alkyl and Aryl Transferases/antagonists & inhibitors , Apoptosis/drug effects , Benzamides/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/physiopathology , Caspases/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Drug Synergism , Farnesyltranstransferase/antagonists & inhibitors , Gene Expression/drug effects , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/pathology , Giant Cell Tumor of Bone/physiopathology , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Pamidronate , Prenylation/drug effects , RNA, Messenger/metabolism , S Phase/drug effects , Tumor Cells, Cultured
9.
J Ethnopharmacol ; 134(1): 130-5, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21130852

ABSTRACT

AIM OF THE STUDY: Astragali radix (AR) is a widely used traditional medicine in oriental countries for treating various diseases including cardiovascular disease (CVD). We investigated the effects of AR extracts on rat cardiomyocytes and H9C2 cardiac cells as well as identified many target genes that mediate the effect of AR. MATERIALS AND METHODS: The effect of AR extracts on cell proliferation was assessed and cDNA microarray technique was used to analyse the differential gene expressions upon AR treatment in cardiac cells. One of the selected target genes was over-expressed to elucidate its role in cell proliferation. RESULTS: AR was shown to promote the proliferation of neonatal rat cardiomyocytes and H9C2 cells. Results of cDNA microarray hybridization showed that N-G,N-G-dimethylarginine dimethylaminohydrolase 1 (DDAH1) gene was up-regulated in AR-treated H9C2 cells and the results were further confirmed by reverse transcription polymerase chain reaction. Over-expression of DDAH1 gene in H9C2 cells significantly enhances the cell proliferation. Moreover, a drastic drop of DDAH1 expression in rat ventricular myocardium was observed from day 3 to day 5 after birth, which is the critical transition of cardiomyocytes from hyperplastic to hypertrophic growth. CONCLUSIONS: AR promotes cardiac cell proliferation and up-regulates the DDAH1, an enzyme that metabolized the endogenous nitric oxide (NO) synthase inhibitor. The effect of AR on the metabolism of NO deserves future investigation.


Subject(s)
Amidohydrolases/metabolism , Cell Proliferation/drug effects , Heart Ventricles/drug effects , Plant Extracts/pharmacology , Animals , Astragalus Plant/chemistry , Base Sequence , DNA Primers , Gene Expression/drug effects , Heart Ventricles/cytology , Heart Ventricles/enzymology , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
10.
BMC Mol Biol ; 10: 101, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19883516

ABSTRACT

BACKGROUND: CITED proteins belong to a family of non-DNA-binding transcriptional co-regulators that are characterized by a conserved ED-rich domain at the C-terminus. This family of genes is involved in the regulation of a variety of transcriptional responses through interactions with the CBP/p300 integrators and various transcription factors. In fish, very little is known about the expression and functions of CITEDs. RESULTS: We have characterized two closely related but distinct CITED3 genes, gcCited3a and gcCited3b, from the hypoxia-tolerant grass carp. The deduced gcCITED3a and gcCITED3b proteins share 72% amino acid identity, and are highly similar to the CITED3 proteins of both chicken and Xenopus. Northern blot analysis indicates that the mRNA expression of gcCited3a and gcCited3b is strongly induced by hypoxia in the kidney and liver, respectively. Luciferase reporter assays demonstrated that both gene promoters are activated by gcHIF-1. Further, ChIP assays comparing normal and hypoxic conditions reveal differential in vivo binding of gcHIF-1 to both gene promoters in kidney and liver tissues. HRE-luciferase reporter assays demonstrated that both gcCITED3a and gcCITED3b proteins inhibit gcHIF-1 transcriptional activity, and GST pull-down assays confirmed that both proteins bind specifically to the CH1 domain of the grass carp p300 protein. CONCLUSION: The grass carp gcCITED3a and gcCITED3b genes are differentially expressed and regulated in different fish organs in response to hypoxic stress. This is the first report demonstrating in vivo regulation of two closely-related CITED3 isogenes by HIF-1, as well as CITED3 regulation of HIF-1 transcriptional activity in fish. Overall, our findings suggest that unique molecular mechanisms operate through these two gcCITED3 isoforms that likely play an important regulatory role in the hypoxic response in the grass carp.


Subject(s)
Adaptation, Physiological , Carps/metabolism , Fish Proteins/chemistry , Fish Proteins/metabolism , Hypoxia/metabolism , Sequence Homology, Amino Acid , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Carps/genetics , Chromatin Immunoprecipitation , Cricetinae , Cricetulus , Fish Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases/metabolism , Molecular Sequence Data , Phylogeny , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Time Factors , Transcriptional Activation/genetics , p300-CBP Transcription Factors/metabolism
11.
Phys Rev Lett ; 102(12): 121602, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19392265

ABSTRACT

We show that hierarchically small vacuum expectation values of the superpotential in supersymmetric theories can be a consequence of an approximate R symmetry. We briefly discuss the role of such small constants in moduli stabilization and understanding the huge hierarchy between the Planck and electroweak scales.

12.
Phys Rev Lett ; 98(18): 181602, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17501559

ABSTRACT

We study possible correlations between properties of the observable and hidden sectors in heterotic string theory. Specifically, we analyze the case of the Z6-II orbifold compactification which produces a significant number of models with the spectrum of the supersymmetric standard model. We find that requiring realistic features does affect the hidden sector such that hidden sector gauge group factors SU(4) and SO(8) are favored. In the context of gaugino condensation, this implies low energy supersymmetry breaking.

13.
Clin Pharmacol Ther ; 81(3): 346-53, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17339864

ABSTRACT

This study explores the hypotheses that: (1) ethanol will interact with dl-Methylphenidate (MPH) to enantioselectively elevate plasma d-MPH, and primarily yield l-ethylphenidate as a transesterification metabolite; (2) women will exhibit lower relative bioavailability of MPH than men; and (3) sex-dependent differences in subjective effects will exist. dl-MPH HCl (0.3 mg/kg) was administered orally 30 min before ethanol, 30 min after ethanol (0.6 gm/kg), or without ethanol, in a randomized, normal subject three-way crossover study of 10 men and 10 women. Pharmacokinetic parameters were compared. Subjective effects were recorded using visual analog scales. One subject was a novel poor MPH metabolizer whose data were analyzed separately. Ethanol after or before MPH significantly (P<0.0001) elevated the geometric mean for the maximum d-MPH plasma concentration (C(max) (+/-SD)) from 15.3 (3.37) ng/ml to 21.5 (6.81) and 21.4 (4.86), respectively, and raised the corresponding geometric mean for the area under the concentration-time curve values from 82.9 (21.7) ng ml/h to 105.2 (23.5) and 102.9 (19.2). l-MPH was present in plasma only at 1-3% of the concentration of d-MPH, except in the poor metabolizer where l-MPH exceeded that of d-MPH. The metabolite l-ethylphenidate frequently exceeded 1 ng/ml in plasma, whereas d-ethylphenidate was detected only in low pg/ml concentrations. Women reported a significantly greater stimulant effect than men when questioned "Do you feel any drug effect?" (P<0.05), in spite of lower mean plasma d-MPH area under the response-time curves in women. Ethanol elevates plasma d-MPH C(max) and area under the concentration-time curve by approximately 40% and 25%, respectively. If the poor metabolizer of MPH proves to be a distinct phenotype, determining the genetic mechanism may be of value for individualizing drug therapy. The more pronounced stimulant effects experienced by women have sex-based abuse liability implications.


Subject(s)
Central Nervous System Depressants/pharmacology , Central Nervous System Depressants/pharmacokinetics , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/pharmacokinetics , Ethanol/pharmacology , Ethanol/pharmacokinetics , Methylphenidate/pharmacology , Methylphenidate/pharmacokinetics , Adult , Area Under Curve , Biotransformation , Blood Pressure/drug effects , Body Temperature/drug effects , Cross-Over Studies , Drug Interactions , Female , Heart Rate/drug effects , Humans , Male , Methylphenidate/analogs & derivatives , Methylphenidate/metabolism , Pharmacogenetics , Respiratory Mechanics/drug effects , Sex Characteristics , Stereoisomerism
15.
BMC Mol Biol ; 7: 27, 2006 Sep 11.
Article in English | MEDLINE | ID: mdl-16961934

ABSTRACT

BACKGROUND: Current understanding on the relationships between hypoxia, hypoxia-inducible factor-1 (HIF-1) and telomerase reverse transcriptase (TERT) gene expression are largely based on in vitro studies in human cancer cells. Although several reports demonstrated HIF-1- mediated upregulation of the human TERT gene under hypoxia, conflicting findings have also been reported. Thus far, it remains uncertain whether these findings can be directly extrapolated to non-tumor tissues in other whole animal systems in vivo. While fish often encounter environmental hypoxia, the in vivo regulation of TERT by hypoxia in non-neoplastic tissues of fish remains virtually unknown. RESULTS: The adult marine medaka (Oryzias melastigma) was employed as a model fish in this study. We have cloned and characterized a 3261-bp full-length TERT cDNA, omTERT, which encodes a protein of 1086 amino acids. It contains all of the functional motifs that are conserved in other vertebrate TERTs. Motif E is the most highly conserved showing 90.9-100% overall identity among the fish TERTs and 63.6% overall identity among vertebrates. Analysis of the 5'-flanking sequence of the omTERT gene identified two HRE (hypoxia-responsive element; nt. - 283 and - 892) cores. Overexpression of the HIF-1alpha induced omTERT promoter activity as demonstrated using transient transfection assays. The omTERT gene is ubiquitously expressed in fish under normoxia, albeit at varying levels, where highest expression was observed in gonads and the lowest in liver. In vivo expression of omTERT was significantly upregulated in testis and liver in response to hypoxia (at 96 h and 48 h, respectively), where concomitant induction of the omHIF-1alpha and erythropoietin (omEpo) genes was also observed. In situ hybridization analysis showed that hypoxic induction of omTERT mRNA was clearly evident in hepatocytes in the caudal region of liver and in spermatogonia-containing cysts in testis. CONCLUSION: This study demonstrates for the first time, hypoxic regulation of TERT expression in vivo in a whole fish system. Our findings support the notion that hypoxia upregulates omTERT expression via omHIF-1 in non-neoplastic fish liver and testis in vivo. Overall, the structure and regulation of the TERT gene is highly conserved in vertebrates from fish to human.


Subject(s)
DNA-Binding Proteins/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia/enzymology , Oryzias/metabolism , Telomerase/biosynthesis , Amino Acid Motifs , Amino Acid Sequence , Animals , Cloning, Molecular , DNA-Binding Proteins/genetics , Enzyme Induction , Eye Proteins/genetics , Female , Humans , Hypoxia/genetics , In Situ Hybridization , Liver/enzymology , Male , Molecular Sequence Data , Muscle Proteins/genetics , Oryzias/genetics , Oxidative Stress , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Spermatozoa/enzymology , Telomerase/genetics , Testis/enzymology , Transcriptional Activation , Vertebrates/genetics , Viscera/enzymology
16.
BMC Mol Biol ; 7: 15, 2006 Apr 20.
Article in English | MEDLINE | ID: mdl-16623959

ABSTRACT

BACKGROUND: Hypoxia-inducible factors (HIFs) are involved in adaptive and survival responses to hypoxic stress in mammals. In fish, very little is known about the functions of HIFs. RESULTS: We have cloned and characterized two distinct HIF-alpha cDNAs--gcHIF-1alpha and gcHIF-4alpha--from the hypoxia-tolerant grass carp. The deduced gcHIF-1alpha protein is highly similar to the HIF-1alphas (57-68%) from various vertebrate species, while gcHIF-4alpha is a novel isoform, and shows an equivalent degree of amino acid identity (41-47%) to the HIF-1alpha, HIF-2alpha and HIF-3alpha proteins so far described. Parsimony analysis indicated that gcHIF-4alpha is most closely related to the HIF-3alpha proteins. Northern blot analysis showed that mRNA levels of gcHIF-1alpha and gcHIF-4alpha differ substantially under normoxic and hypoxic conditions, while Western blot studies demonstrated that the endogenous protein levels for both gcHIF-1alpha and gcHIF-4alpha are similarly responsive to hypoxia. Our findings suggest that both gcHIF-1alpha and gcHIF-4alpha are differentially regulated at the transcriptional and translational levels. HRE-luciferase reporter assays show that both proteins function as transcription activators and play distinct roles in modulating the hypoxic response in grass carp. CONCLUSION: There are at least two distinct HIF-alpha isoforms--gcHIF-1alpha and gcHIF-4alpha--in the hypoxia-tolerant grass carp, which are differentially expressed and regulated in different fish organs in response to hypoxic stress. Overall, the results suggest that unique molecular mechanisms operate through these two HIF-alpha isoforms, which underpin the hypoxic response in the hypoxia-tolerant grass carp.


Subject(s)
Carps/physiology , Gene Expression Regulation/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern/veterinary , Blotting, Western/veterinary , CHO Cells , Carps/classification , Carps/genetics , Cell Line , Cloning, Molecular/methods , Cricetinae , Cricetulus , DNA Primers/chemistry , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Gene Expression Regulation/genetics , Gene Order , Hypoxia/metabolism , Hypoxia/veterinary , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver/physiology , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary
17.
Clin Pharmacokinet ; 40(10): 753-72, 2001.
Article in English | MEDLINE | ID: mdl-11707061

ABSTRACT

The psychostimulants methylphenidate, amphetamine and pemoline are among the most common medications used today in child and adolescent psychiatry for the treatment of patients with attention-deficit hyperactivity disorder. Frequently, these medications are used in combination with other medications on a short or long term basis. The present review examines psychostimulant pharmacology, summarises reported drug-drug interactions and explores underlying pharmacokinetic and pharmacodynamic considerations for interactions. A computerised search was undertaken using Medline (1966 to 2000) and Current Contents to provide the literature base for reports of drug-drug interactions involving psychostimulants. These leads were further cross-referenced for completeness of the survey. Methylphenidate appears to be more often implicated in pharmacokinetic interactions suggestive of possible metabolic inhibition, although the mechanisms still remain unclear. Amphetamine was more often involved in apparent pharmacodynamic interactions and could potentially be influenced by medications affecting cytochrome P450 (CYP) 2D6. No published reports of drug interactions involving pemoline were found. The alpha2-adrenergic agonists clonidine and guanfacine have been implicated in several interactions. Perhaps best documented is their antagonism by tricyclic antidepressants and phenothiazines. In additional, concurrent beta-blocker use, or abrupt discontinuation, can lead to hypertensive response. Although there are few published well-controlled interaction studies with psychostimulants and alpha2-adrenergic agonists, it appears that these agents may be safely coadministered. The interactions of monoamine oxidase inhibitors with psychostimulants represent one of the few strict contraindications.


Subject(s)
Adrenergic alpha-Agonists , Anticonvulsants , Antidepressive Agents , Antihypertensive Agents , Attention Deficit Disorder with Hyperactivity , Central Nervous System Stimulants , Adrenergic alpha-Agonists/pharmacokinetics , Adrenergic alpha-Agonists/therapeutic use , Adult , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/therapeutic use , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/metabolism , Central Nervous System Stimulants/pharmacokinetics , Central Nervous System Stimulants/therapeutic use , Child , Drug Interactions , Humans
19.
Pharm Res ; 17(4): 381-4, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10870979

ABSTRACT

PURPOSE: To determine the relative bioavailability of two marketed, immediate-release methylphenidate tablets. The study used a replicated study design to characterize intrasubject variability, and determine bioequivalence using both average and individual bioequivalence criteria. METHODS: A replicated crossover design was employed using 20 subjects. Each subject received a single 20 mg dose of the reference tablet on two occasions and two doses of the test tablet on two occasions. Blood samples were obtained for 10 hr after dosing, and plasma was assayed for methylphenidate by GC/MS. RESULTS: The test product was more rapidly dissolved in vitro and more rapidly absorbed in vivo than the reference product. The mean Cmax and AUC(0-infinity) differed by 11% and 9%, respectively. Using an average bioequivalence criterion, the 90% confidence limits for the Ln-transformed Cmax and AUC(0-infinity), comparing the two replicates of the test to the reference product, fell within the acceptable range of 80-125%. Using an individual bioequivalence criterion the test product failed to demonstrate equivalence in Cmax to the reference product. CONCLUSIONS: The test and reference tablets were bioequivalent using an average bioequivalence criterion. The intrasubject variability of the generic product was greater and the subject-by-formulation interaction variance was borderline high. For these reasons, the test tablets were not individually bioequivalent to the reference tablets.


Subject(s)
Methylphenidate/pharmacokinetics , Adult , Area Under Curve , Cross-Over Studies , Gas Chromatography-Mass Spectrometry , Humans , Male , Methylphenidate/administration & dosage , Methylphenidate/blood , Tablets , Therapeutic Equivalency , United States , United States Food and Drug Administration
20.
Drug Metab Dispos ; 28(6): 620-4, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10820132

ABSTRACT

Ethylphenidate was recently reported as a novel drug metabolite in two overdose fatalities where there was evidence of methylphenidate and ethanol coingestion. This study explores the pharmacokinetics of ethylphenidate relative to methylphenidate and the major metabolite ritalinic acid, in six healthy subjects who received methylphenidate and ethanol under controlled conditions. Subjects (three males, three females) received a single oral dose of methylphenidate (20 mg; two 10-mg tablets) followed by consumption of ethanol (0.6 g/kg) 30 min later. Methylphenidate, ritalinic acid, and ethylphenidate were quantified using liquid chromatography-tandem mass spectrometry. Ethylphenidate was detectable in the plasma and urine of all subjects after ethanol ingestion. The mean (+/-S.D.) area under the concentration versus time curve for ethylphenidate was 1.2 +/- 0.7 ng/ml/h, representing 2.3 +/- 1.3% that of methylphenidate (48 +/- 12 ng/ml/h). A significant correlation was observed between the area under the concentration versus time curve of methylphenidate and that of ethylphenidate. In view of the known dopaminergic activity of racemic ethylphenidate, it remains possible that under certain circumstances of higher level dosing, e.g., in the abuse of methylphenidate and ethanol, the metabolite ethylphenidate may contribute to drug effects.


Subject(s)
Alcohol Drinking/metabolism , Ethanol/metabolism , Methylphenidate/analogs & derivatives , Methylphenidate/metabolism , Adult , Alcohol Drinking/adverse effects , Analysis of Variance , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/metabolism , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Female , Humans , Male , Methylphenidate/administration & dosage , Methylphenidate/blood , Methylphenidate/toxicity , Methylphenidate/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...