Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
CPT Pharmacometrics Syst Pharmacol ; 3: e112, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24785551

ABSTRACT

Optimizing anticancer therapeutics needs to account for variable drug responses in heterogeneous cell populations within the tumor as well as in organs of toxicity. To address cell heterogeneity, we propose a multiscale modeling approach-from in vitro to preclinical and clinical studies-to develop cell-type-specific pharmacokinetic-pharmacodynamic (PK-PD) models. A physiologically based mechanistic modeling approach integrating data from aqueous solutions, U87 glioma cells, mice, and cancer patients was utilized to characterize the brain disposition of temozolomide (TMZ), the cornerstone of chemotherapy against glioblastoma multiforme. The final model represented intracellular normal brain and brain tumor compartments in which TMZ pH-dependent conversion to the DNA-alkylating species leads to the formation of DNA adducts that serve as an entry point for a PD model. This multiscale protocol can be extended to account for TMZ PK-PD in different cell populations, thus providing a critical tool to personalize TMZ-based chemotherapy on a cell-type-specific basis.

2.
Article in English | MEDLINE | ID: mdl-24429593

ABSTRACT

A unified approach to optimize multidrug chemotherapy using a pharmacokinetic (PK)/enhanced pharmacodynamic model was developed using the vascular endothelial growth factor receptor (VEGFR) signaling system. The base VEGFR network model, characterized by ligand-receptor interactions, enzyme recruitment (Grb2-Sos, phospholipase C γ (PLCγ), and phosphoinositide-3 kinase (PI3K)), and downstream mitogen-activated protein kinase and Akt cascade activation, was linked to a sunitinib (VEGFR inhibitor) PK model and underwent Sobol sensitivity analysis that revealed potential sunitinib-enhancing mechanisms. Drugs targeting these mechanisms (a VEGF inhibitor, a PI3K inhibitor, a PLCγ inhibitor, and a mitogen-activated protein kinase inhibitor) and sunitinib were input to optimization-based control analyses to design multidrug regimens that maintained 80% pERK and pAkt inhibition for 28 days while minimizing drug dose. The resultant combination regimens contained both continuous and discontinuous schedules, mostly at low doses, and were altered by oncogenic mutations. This pipeline of computational analyses demonstrates how model-based methods can capture the complexities of drug action, tailor cancer chemotherapy, and empower personalized medicine.

4.
Article in English | MEDLINE | ID: mdl-24448020

ABSTRACT

Declining success rates coupled with increased costs is leading to an inevitable breaking point in the drug development pipeline. Can we avoid it by incorporating the vast mechanistic understanding of drug action? A recent review highlights this dilemma and proposes "quantitative logic gate" modeling as a solution.(1) The goal of this commentary is to contrast this approach with mechanistic biochemical network models, which, although alluded to by Kiruoac and Onsum, requires a closer analysis.

5.
CNS Neurol Disord Drug Targets ; 9(4): 403-28, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20522014

ABSTRACT

Tauopathies, including Alzheimer's disease, are neurodegenerative diseases characterized by the deposition of hyperphosphorylated tau protein in the central nervous system, and are the major cause of dementia in later life. Considerable advances have been made in developing mouse models that recapitulate, to varying extents, the development of human tau pathology, and the learning and memory deficits characteristic of some tauopathies. Furthermore, such models have been used to show promising disease-modifying effects in pre-clinical testing of new therapeutics. Various strategies have been utilised to generate mouse models of tauopathies. Some of the most enlightening models developed to date either constitutively or inducibly express pathogenic tau mutations. These animals have been instrumental in defining critical disease-related mechanisms, including the observation that tangles are not the toxic form of tau in disease. Here, we discuss the strengths and weaknesses of well characterised transgenic models that emulate human tauopathy, and include a comprehensive listing of the main phenotypic characteristics of all reported tau transgenic rodents. We summarise the use of tau mice for the development and evaluation of new therapeutic approaches, and their utility in identifying novel drug targets. In addition, we review the parameters to be considered in the development of the next generation of mouse models of tauopathy, aimed at further increasing our understanding of disease aetiology and in evaluating novel treatments.


Subject(s)
Disease Models, Animal , Drug Discovery/methods , Mice, Transgenic , Tauopathies/drug therapy , Animals , Humans , Mice , Mice, Transgenic/genetics , Tauopathies/genetics , tau Proteins/drug effects , tau Proteins/genetics , tau Proteins/physiology
6.
Cell Mol Life Sci ; 64(13): 1701-14, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17453144

ABSTRACT

Tauopathies are a group of neurodegenerative diseases characterised by intracellular deposits of the microtubule-associated protein tau. The most typical example of a tauopathy is Alzheimer's disease. The importance of tau in neuronal dysfunction and degeneration has been demonstrated by the discovery of dominant mutations in the MAPT gene, encoding tau, in some rare dementias. Recent developments have shed light on the significance of tau phosphorylation and aggregation in pathogenesis. Furthermore, emerging evidence reveals the central role played by tau pre-mRNA processing in tauopathies. The present review focuses on the current understanding of tau-dependent pathogenic mechanisms and how realistic therapies for tauopathies can be developed.


Subject(s)
RNA/metabolism , Tauopathies/metabolism , Tauopathies/therapy , tau Proteins/metabolism , Alternative Splicing/genetics , Animals , Humans , Phosphorylation , Protein Conformation , tau Proteins/chemistry , tau Proteins/genetics
7.
Mol Ther ; 15(6): 1100-5, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17426712

ABSTRACT

We studied the ability of heat shock, DnaJ-like-1 (HSJ1) proteins (which contain DnaJ and ubiquitin-interacting motifs) to reduce polyglutamine-mediated inclusion formation. The experiments demonstrated that expression of heat shock protein 70 (hsp70), hsp40, HSJ1a, and HSJ1b significantly reduced protein inclusion formation in a model of spinal and bulbar muscular atrophy (SBMA). HSJ1a also mediated a significant decrease in the number of inclusions formed in a primary neuronal model of protein aggregation. Studies to elucidate the mechanisms underlying these reductions showed that hsp70 and hsp40 increased chaperone-mediated refolding. In contrast, expression of HSJ1 proteins did not promote chaperone activity but caused an increase in ubiquitylation. Furthermore, HSJ1a was associated with a ubiquitylated luciferase complex, and in the presence of HSJ1a but not an HSJ1a UIM mutant (HSJ1a-deltaUIM) there was a reduction in luciferase protein levels. Together these results show that HSJ1 proteins mediated an increase in target protein degradation via the ubiquitin-proteasome system (UPS). We also found that the expression of HSJ1a significantly decreased the number of neurons containing inclusions in an in vivo model of polyglutamine disease. These findings indicate that targeted modification of the UPS to facilitate degradation of misfolded proteins may represent a highly effective therapeutic avenue for the treatment of polyglutamine disease.


Subject(s)
HSP40 Heat-Shock Proteins/genetics , Inclusion Bodies/metabolism , Muscular Atrophy, Spinal/therapy , Peptides/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Dependovirus/genetics , Genetic Vectors/genetics , HSP40 Heat-Shock Proteins/physiology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/physiology , Immunohistochemistry , Immunoprecipitation , Luciferases/chemistry , Luciferases/genetics , Luciferases/metabolism , Male , Microscopy, Fluorescence , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Neurons/metabolism , Peptides/genetics , Protein Folding , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Transfection
8.
Mol Ther ; 15(6): 1100-1105, 2007 Jun.
Article in English | MEDLINE | ID: mdl-28182921

ABSTRACT

We studied the ability of heat shock, DnaJ-like-1 (HSJ1) proteins (which contain DnaJ and ubiquitin-interacting motifs) to reduce polyglutamine-mediated inclusion formation. The experiments demonstrated that expression of heat shock protein 70 (hsp70), hsp40, HSJ1a, and HSJ1b significantly reduced protein inclusion formation in a model of spinal and bulbar muscular atrophy (SBMA). HSJ1a also mediated a significant decrease in the number of inclusions formed in a primary neuronal model of protein aggregation. Studies to elucidate the mechanisms underlying these reductions showed that hsp70 and hsp40 increased chaperone-mediated refolding. In contrast, expression of HSJ1 proteins did not promote chaperone activity but caused an increase in ubiquitylation. Furthermore, HSJ1a was associated with a ubiquitylated luciferase complex, and in the presence of HSJ1a but not an HSJ1a UIM mutant (HSJ1a-ΔUIM) there was a reduction in luciferase protein levels. Together these results show that HSJ1 proteins mediated an increase in target protein degradation via the ubiquitin-proteasome system (UPS). We also found that the expression of HSJ1a significantly decreased the number of neurons containing inclusions in an in vivo model of polyglutamine disease. These findings indicate that targeted modification of the UPS to facilitate degradation of misfolded proteins may represent a highly effective therapeutic avenue for the treatment of polyglutamine disease.

9.
Oncogene ; 25(6): 954-8, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16247462

ABSTRACT

A second generation genetically-engineered cell-based drug delivery system, referred to as apoptotic-induced drug delivery (AIDD), was developed using endothelial cells (ECs) that undergo apoptosis upon binding of vascular endothelial growth factor (VEGF) to a Flk-1:Fas fusion protein (FF). This new AIDD was redesigned using mouse brain derived ECs, bEnd3 cells, and an adenovirus vector in order to enhance and control the expression of FF. The FF was tagged with a HA epitope (FFHA) and designed to be coexpressed with green fluorescence protein (GFP) by the regulation of cytomegalovirus promoters in the adenovirus vector. bEnd3 cells showed favorable coexpression of FFHA and GFP consistent with the multiplicity of infection of the adenovirus. Immunofluorescence analysis demonstrated that FFHA was localized at the plasma membrane, whereas GFP was predominantly located in the cytoplasm of ECs. Cell death was induced by VEGF, but not by platelet derived growth factor or fibroblast growth factor in a dose-dependent manner (range 2-20 ng/ml), and revealed caspase-dependent apoptotic profiles. The FFHA expressing bEnd3 cells underwent apoptosis when cocultured with a glioma cell (SF188V+) line able to overexpress VEGF. The combined data indicated that the FFHA adenovirus system can induce apoptotic signaling in ECs in response to VEGF, and thus, is an instrumental modification to the development of AIDD.


Subject(s)
Apoptosis/drug effects , Brain/cytology , Drug Delivery Systems/methods , Vascular Endothelial Growth Factor A/pharmacology , Adenoviridae/genetics , Animals , Apoptosis/genetics , Base Sequence , Cells, Cultured , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/pathology , Coculture Techniques , Endothelial Cells/virology , Epitopes/genetics , Epitopes/metabolism , Genetic Vectors/genetics , Glioma/drug therapy , Glioma/pathology , Humans , Mice , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , fas Receptor
10.
Brain Res Mol Brain Res ; 137(1-2): 104-9, 2005 Jun 13.
Article in English | MEDLINE | ID: mdl-15950767

ABSTRACT

A number of neurodegenerative diseases, including Alzheimer's disease (AD), are characterized by intraneuronal accumulation of the tau protein. Some forms of FTDP-17 are caused by mutations in the tau gene affecting exon 10 splicing. Therefore, dysregulation of tau pre-mRNA splicing may be a contributing factor to sporadic tauopathies. To address this question, we devised a real-time RT-PCR strategy based on the use of a single fluorogenic probe to evaluate the ratio between tau isoforms containing or lacking exon 10 (4R/3R ratio) in post-mortem brain samples. We found a two- to six-fold increase in the 4R/3R ratio in cases of FTDP-17 linked to a splice site mutation, hence confirming the validity of the strategy. The difference in the 4R/3R ratio in the superior temporal and superior frontal gyri between AD and control brains was not statistically significant. Similarly, there was no significant difference in the 4R/3R ratio between Pick's disease cases and controls, indicating that the predominance of tau3R protein in PiD reflects post-translational modifications of specific isoforms. This study indicates that post-translational events are likely to be the main factors controlling tau isoform composition in sporadic tauopathies and highlights the benefit of quantitative RT-PCR in the assessment of splicing abnormalities in tauopathies.


Subject(s)
Alternative Splicing/genetics , Brain/metabolism , Mutation/genetics , Polymorphism, Genetic/genetics , Tauopathies/genetics , tau Proteins/genetics , Aged , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Base Sequence/genetics , Brain/pathology , Brain/physiopathology , Dementia/genetics , Dementia/metabolism , Dementia/physiopathology , Exons/genetics , Humans , Middle Aged , Molecular Sequence Data , Pick Disease of the Brain/genetics , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/physiopathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Processing, Post-Translational/genetics , RNA Splice Sites/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Tauopathies/metabolism , Tauopathies/physiopathology , tau Proteins/metabolism
11.
Neurosci Lett ; 354(1): 54-8, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14698481

ABSTRACT

Expansion of the polyglutamine repeat region of the androgen receptor (AR) results in Kennedy's disease, a neurological disorder typified by degeneration of motor neurons in the brain stem and spinal cord. As the AR has been shown to inhibit beta-catenin dependent (Wnt) signalling we asked if expansion of the polyglutamine repeats might affect this property of the protein. Using the TOPflash/FOPflash reporter assay we found that a pathogenic form of the AR containing 51 glutamine repeats showed a consistent, though minimal, reduction in its ability to inhibit beta-catenin-mediated transcription, in comparison to a non-pathogenic form with 20 repeats. A reduced ability to inhibit Wnt signalling may thus contribute in part to the underlying aetiology of Kennedy's disease.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA Repeat Expansion , DNA-Binding Proteins/metabolism , Muscular Atrophy, Spinal/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Zebrafish Proteins , Cell Line , Genes, Reporter , Humans , Kidney/cytology , Lymphoid Enhancer-Binding Factor 1 , Muscular Atrophy, Spinal/physiopathology , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Peptides/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Transcription, Genetic/physiology , Wnt Proteins , beta Catenin
12.
Exp Biol Med (Maywood) ; 228(8): 982-90, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12968071

ABSTRACT

The pathogenesis of X-linked spinal and bulbar muscular atrophy (SBMA) has been traced to an expansion of repeated glutamine (Gln) residues within the amino terminus of the human androgen receptor (AR). To examine the mechanisms by which these expanded repeat ARs (Exp-ARs) are toxic to neurons, we have established and characterized a cell culture model by stably transfecting SH-SY 5Y neuroblastoma cells with cDNAs containing either normal AR (81 series; 23 Glns) or Exp-AR (902 series; 56 Glns). At a low passage number, no differences in cell morphology, growth properties, or susceptibility to toxic insults were observed between clones expressing normal AR or Exp-AR. Initially, both types of cultures were found to express similar levels of specific hormone binding in monolayer binding assays. Immunohistochemical studies demonstrated the vast majority of both the normal AR and Exp-AR were localized to the nucleus in the absence and presence of androgen. As the 902 series of clones were propagated, the Exp-AR content in the cells appeared to decline progressively. However, this decrease actually reflects a gradual disappearance of the Exp-AR cell population. No such selection occurred during the propagation of cells expressing the normal AR. This selection against cells expressing physiological levels of Exp-AR occurs in the absence of intracellular aggregates and suggests that mechanisms other than those involving the formation of aggregates underlie the observed toxicity of Exp-ARs.


Subject(s)
Neurons/metabolism , Receptors, Androgen/biosynthesis , Trinucleotide Repeat Expansion/physiology , Apoptosis , Binding, Competitive , Humans , Immunoblotting , Immunohistochemistry , Microscopy, Fluorescence , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , Neurons/pathology , Neurons/ultrastructure , Peptides/genetics , Receptors, Androgen/genetics , Time Factors , Transfection , Tumor Cells, Cultured
13.
Rev. ortop. traumatol. (Madr., Ed. impr.) ; 45(6): 483-486, dic. 2001. ilus
Article in Es | IBECS | ID: ibc-4747

ABSTRACT

Se realiza un estudio retrospectivo de los resultados clínicos y radiológicos de 6 pacientes afectos de escápula elevada congénita (deformidad de Sprengel) tras el tratamiento quirúrgico de los mismos empleando la técnica de Woodward. El nivel medio de elevación prequirúrgica del borde inferior de la escápula respecto a la contralateral fue de 3,3 cm, mientras que el déficit medio de abducción fue de 45°. Con un seguimiento medio de 6 años la elevación del borde inferior escapular había pasado a 1,4 cm y la ganancia media de abducción había mejorado 30° respecto a los valores prequirúrgicos. En 2 casos se realizó una osteotomía clavicular para evitar una posible lesión del plexo braquial al descender la escápula. Debido a la hipoplasia escapular asociada, con una disminución en la longitud vertical, no se debe intentar durante la cirugía llevar el ángulo inferior escapular al mismo nivel del contralateral (AU)


Subject(s)
Female , Child, Preschool , Male , Child , Humans , Scapula/surgery , Limb Deformities, Congenital/surgery , Bone Diseases/surgery , Osteotomy/methods , Retrospective Studies , Treatment Outcome , Follow-Up Studies
14.
Brain Res Bull ; 56(3-4): 209-14, 2001.
Article in English | MEDLINE | ID: mdl-11719252

ABSTRACT

Two definite genetic causes of adult motor neuron degeneration have been identified to date: CAG repeat expansion in the androgen receptor gene in Kennedy's disease and point mutations in the SOD1 gene, encoding the enzyme, Cu/Zn superoxide dismutase, in some familial forms of amyotrophic lateral sclerosis. Although both have unrelated genetic causes, Kennedy's disease and SOD1-linked amyotrophic lateral sclerosis share several pathogenic features. First, expanded androgen receptor and mutant Cu/Zn superoxide dismutase have a propensity to aggregate into insoluble complexes and form inclusion bodies in affected neurons. Deposits of mutant proteins could be detrimental to neuronal viability by interfering with the normal housekeeping functions of chaperones and of the ubiquitin/proteasome system. Secondly, cytoskeletal function may be impaired in both diseases as decreased transactivational activity of expanded androgen receptor may cause an abnormal pattern of tubulin expression in motor neurons in Kennedy's disease and disruption of neurofilament organisation is a hallmark of amyotrophic lateral sclerosis. The concept of activation of overlapping cell death cascades by two distinct genetic defects could help elucidating downstream pathogenic processes and may provide novel targets for pharmacological intervention or gene therapy for the treatment of motor neuron disorders.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Muscular Atrophy, Spinal/classification , Muscular Atrophy, Spinal/genetics , Humans , Neurofilament Proteins/genetics , Peptides/genetics , Receptors, Androgen/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Tubulin/genetics
15.
Cancer Chemother Pharmacol ; 48(2): 169-76, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11561783

ABSTRACT

PURPOSE: Peripheral benzodiazepine receptors (PBRs) are overexpressed in brain tumors compared to normal brain, and could serve as a target to selectively increase anticancer drug delivery through a PBR ligand-drug conjugate system. We have previously synthesized PBR ligand-gemcitabine conjugates based on the model PBR ligand, PK11195. The goal of the current study was to examine this new drug delivery strategy in an intracerebral xenograft model by measurement of steady-state drug distribution following administration of gemcitabine (GEM) and PK11195-GEM. METHODS: In vitro PBR receptor binding and cytotoxicity assays were used to screen three different PK11195-GEM conjugates (GG01, GG02, GG03) in human SF126 glioma cells. Based on these findings and the favorable chemical stability of GG01, here referred as PK11195-GEM, pharmacokinetic investigations of PK11195-GEM and GEM were conducted in male rats. These studies consisted of single-dose and steady-state dosing regimen studies, the latter to assess drug distribution in normal brain and brain tumors. PK11195-GEM and GEM were measured in blood and tissue samples by HPLC. RESULTS: All PBR-GEM conjugates demonstrated appreciable receptor binding affinity and cytotoxicity with mean IC50 values ranging from 248 to 376 nM and 5.6 to 29.1 nM, respectively. The cytotoxicity of GEM was comparable with a mean IC50 value of 5.9 nM. Following administration of single 8 mg/kg doses of PK11195-GEM to rats (n=4), PK11195-GEM had a mean total clearance of 126.3 +/- 29.6 ml/min per kg, and a volume of distribution at steady-state of 1,261.9 +/- 31.05 ml/kg that resulted in a very short elimination half-life of 16.1 +/- 5.8 min. In comparison GEM had a similar volume of distribution (993.8 +/- 131.6 ml/kg), reduced clearance (3.4 +/- 0.8 ml/ min per kg), and longer half-life (235.6 +/- 26.7 min). In nude rats bearing intracerebral tumors, mean steady-state tumor/plasma, tumor/right brain, and tumor/left brain PK 11195-GEM concentration ratios were 1.75 +/- 0.46, 5.49 +/- 5.2, and 9.96 +/- 3.2, respectively. The analogous values following GEM administrations were 0.81 0.5, 3.67 1.57, and 5.21 +/- 1.95, respectively. These values indicate a minimum twofold increase in tumor target selectivity for the conjugate delivery system compared to GEM treatment. CONCLUSION: Targeting intracellular PBRs is a new drug delivery strategy based on the use of low molecular weight drug conjugates that can be administered systemically. It was demonstrated under steady-state conditions that PK11195-GEM possessed a twofold enhancement in brain tumor selectivity compared to GEM alone. This type of target selectivity would allow higher tumor concentrations to be achieved in conjunction with lower drug concentrations in normal or non-target tissues.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/administration & dosage , Drug Delivery Systems , Isoquinolines/administration & dosage , Receptors, GABA-A/metabolism , Animals , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/toxicity , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/metabolism , Deoxycytidine/pharmacokinetics , Deoxycytidine/toxicity , Glioma/drug therapy , Glioma/metabolism , Humans , Isoquinolines/metabolism , Isoquinolines/pharmacokinetics , Isoquinolines/toxicity , Male , Rats , Rats, Nude , Rats, Sprague-Dawley , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
16.
J Neurochem ; 78(4): 854-61, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11520906

ABSTRACT

Androgens regulate the physiology of motor neurones both during development and in adult life. In particular, androgens increase the rate of axonal regeneration after axotomy, an effect correlated with the up-regulation of tubulin. In order to determine whether this was the result of a direct hormone action on neurones, we examined the effect of testosterone on microtubular proteins in human neuroblastoma SH-SY5Y cells. Treatment of proliferating SH-SY5Y cells with testosterone resulted in an up-regulation of alpha- and beta-tubulin. By contrast, no change in tubulin was observed either in cells differentiated into a neuronal phenotype by retinoic acid or in adrenal SW13 cells. We also show that an up-regulation of the ubiquitous beta(II)-tubulin and of the neurone-specific beta(III)-tubulin isoforms contributes to the overall increase in tubulin in response to androgen treatment. The increase in tubulin levels following testosterone treatment was abolished by co-incubation with antiandrogens, indicating that this effect is mediated through a classical mechanism of steroid action. The two microtubule-associated proteins, tau and MAP2b, remained unchanged following testosterone exposure. Thus, these results demonstrate that tubulin is a direct neuronal target of androgen regulation and suggest that dysregulation of tubulin expression may contribute to the pathogenesis of some motor neuronopathies.


Subject(s)
Microtubules/drug effects , Neurons/metabolism , Testosterone/pharmacology , Tubulin/metabolism , Androgen Antagonists/pharmacology , Animals , Blotting, Northern , Brain Chemistry , Cell Differentiation/physiology , Cyproterone Acetate/pharmacology , Flutamide/pharmacology , Humans , Immunoblotting , Liver/chemistry , Mice , Microtubules/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Isoforms , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Tumor Cells, Cultured , tau Proteins/metabolism
17.
Biochem Soc Symp ; (67): 73-80, 2001.
Article in English | MEDLINE | ID: mdl-11447841

ABSTRACT

The microtubule-associated protein, tau, is the principal component of paired helical filaments (PHFs) in Alzheimer's disease. PHF-tau is highly phosphorylated and a total of 25 sites of phosphorylation have so far been identified. Many of these sites are serine or threonine residues that are immediately followed in the sequence by proline residues, and hence are candidate phosphorylation sites for proline-directed kinases. In vitro, glycogen synthase kinase-3 (GSK-3), extracellular signal-related kinase-1 and -2, and mitogen-activated protein kinases, p38 kinase and c-jun N-terminal kinase, all phosphorylate many of these sites, although with different efficiencies for particular sites. Phosphorylation studies in transfected cells and neurons show that GSK-3 phosphorylates tau more extensively than do these other proline-directed kinases. Mutations in tau have been shown to affect in vitro phosphorylation of tau by GSK-3. The Arg406-->Trp (R406W) tau mutation also affects tau phosphorylation in cells.


Subject(s)
tau Proteins/metabolism , Alzheimer Disease/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , In Vitro Techniques , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 10 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutation , Neurons/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases , tau Proteins/chemistry , tau Proteins/genetics
18.
Cancer Res ; 61(14): 5491-8, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454697

ABSTRACT

The angiogenic phenotype is associated with increased tumor neovascularization and a state of vascular hyperpermeability to macromolecules. Angiogenesis inhibitors could reverse these processes, resulting in tumor capillaries that have normal membrane permeability. It was proposed that the switch from a hyperpermeable to a normal permeable state could have the untoward effect of decreasing tumor concentrations of anticancer drugs coadministered with angiogenesis inhibitors. The current investigation evaluated a potential drug interaction between the angiogenesis inhibitor O-(N-chloroacetyl-carbamoyl)-fumagillol (TNP-470) and the alkylating agent temozolomide (TMZ), in xenograft models that differentially expressed vascular endothelial growth factor (VEGF), a driving force for angiogenesis. Nude rats bearing either s.c. low VEGF (V-) or high VEGF (V+) or intracerebral V+ gliomas were administered either a multiple-dose regimen of TNP-470 or vehicle control. One day after the last dose of vehicle or TNP-470, a steady-state dosing regimen of TMZ was administered with subsequent collection and high-performance liquid chromatography analysis of plasma and either tumor homogenate or tumor microdialysis steady-state TMZ concentrations, and in some cases [5-(3-methyltriazen-1-yl)imidazole-4-carboximide] MTIC, its active metabolite. Microvessel density (MVD) was quantitated by image analysis using an anti-CD31 method. Statistical analyses of pharmacokinetic and pharmacodynamic end points in the control and TNP-470 treatment groups were completed by nonparametric tests. In both the s.c. and intracerebral V+ models, TNP-470 treatment produced significant reductions in TMZ tumor concentrations and tumor:plasma concentration ratios compared with control, being reduced an average of 25% and 50% in the s.c. and intracerebral tumors, respectively. MTIC concentrations in V+ s.c. tumors also were reduced by 50% in the presence of TNP-470. Consistent with the lower extent of neovascularization in the V- tumors, TMZ and MTIC tumor concentrations were not different in TNP-470 and control treatment groups in s.c. tumors. MVD was reduced by TNP-470 compared with vehicle control in the V+ tumors, but was unaltered in V- tumors, attesting to the use of MVD as a pharmacodynamic end point and the effectiveness of TNP-470 as an angiogenesis inhibitor. Angiogenesis inhibitor's pharmacodynamic actions on tumor angiogenesis can produce a reduction in tumor concentrations of coadministered anticancer agents. It is increasingly important to understand the pharmacokinetic and pharmacodynamic behavior of each class of drug so that optimal dosing regimens can be designed.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Alkylating/pharmacokinetics , Dacarbazine/pharmacokinetics , Sesquiterpenes/pharmacology , Animals , Antineoplastic Agents, Alkylating/blood , Antineoplastic Agents, Alkylating/metabolism , Cyclohexanes , Dacarbazine/analogs & derivatives , Dacarbazine/blood , Dacarbazine/metabolism , Dialysis Solutions/chemistry , Dialysis Solutions/metabolism , Drug Interactions , Humans , Male , Microdialysis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , O-(Chloroacetylcarbamoyl)fumagillol , Rats , Rats, Nude , Temozolomide , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
J Clin Oncol ; 19(4): 1183-94, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11181685

ABSTRACT

PURPOSE: To determine the safety and feasibility of delivering multiple cycles of front-line high-dose carboplatin, paclitaxel, and topotecan with peripheral-blood stem-cell (PBSC) support. PATIENTS AND METHODS: Patients were required to have a malignant solid tumor for which they had received no prior chemotherapy. Mobilization of PBSC was achieved with either filgrastim alone or in combination with cyclophosphamide and paclitaxel. Patients then received three or four cycles of high-dose carboplatin (area under the concentration-time curve [AUC] 16), paclitaxel (250 mg/m(2)), and topotecan (10-15 mg/m(2)), with the latter two agents administered as 24-hour infusions and supported with PBSC and filgrastim. Cycles were repeated every 28 days. RESULTS: Twenty patients were enrolled onto the trial and were assessable for toxicity and clinical outcome. Dose-limiting toxicities were stomatitis and prolonged hematopoietic recovery. The maximum-tolerated dose of topotecan was 12.5 mg/m(2) when given with high-dose carboplatin and paclitaxel for three cycles. Four cycles were able to be given with a dose of topotecan of 10 mg/m(2). The pharmacokinetics of each compound were not affected by the other agents. Eleven (85%) of 13 patients with assessable disease responded. CONCLUSION: Multiple cycles of high-dose carboplatin, paclitaxel, and topotecan can be safely administered with filgrastim and PBSC support. The recommended doses for phase II study are carboplatin AUC 16, paclitaxel 250 mg/m(2), and topotecan 10 mg/m(2). Trials are currently being conducted with this regimen as front-line treatment in patients with advanced ovarian cancer and extensive small-cell carcinoma. This approach remains experimental and should be used only in the context of a clinical trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Hematopoietic Stem Cell Transplantation , Neoplasms/therapy , Paclitaxel/administration & dosage , Topotecan/administration & dosage , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Drug Administration Schedule , Female , Hospitalization , Humans , Male , Middle Aged , Neoplasms/drug therapy , Neutropenia/chemically induced , Stomatitis/chemically induced
20.
Adv Drug Deliv Rev ; 45(2-3): 243-53, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11108977

ABSTRACT

Microdialysis is an in vivo sampling technique that was initially developed to measure endogenous substances in the field of neurotransmitter research. In the past decade, microdialysis has been increasingly applied to study the pharmacokinetics and drug metabolism in the blood and various tissues of both animals and humans. This paper describes the general aspects of this in vivo sampling technique followed by the survey of the recent papers regarding the application of microdialysis to characterize anticancer drug disposition in solid tumors. It can be concluded that microdialysis is a very suitable method to obtain drug concentration-time profiles in the interstitial fluid of solid tumors as well as of other variety of tissues.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Microdialysis , Neoplasms/metabolism , Animals , Calibration , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...