Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(9): e0138616, 2015.
Article in English | MEDLINE | ID: mdl-26398286

ABSTRACT

Cell cycle checkpoint intervention is an effective therapeutic strategy for cancer when applied to patients predisposed to respond and the treatment is well-tolerated. A critical cell cycle process that could be targeted is the mitotic checkpoint (spindle assembly checkpoint) which governs the metaphase-to-anaphase transition and insures proper chromosomal segregation. The mitotic checkpoint kinase Mps1 was selected to explore whether enhancement in genomic instability is a viable therapeutic strategy. The basal-a subset of triple-negative breast cancer was chosen as a model system because it has a higher incidence of chromosomal instability and Mps1 expression is up-regulated. Depletion of Mps1 reduces tumor cell viability relative to normal cells. Highly selective, extremely potent Mps1 kinase inhibitors were created to investigate the roles of Mps1 catalytic activity in tumor cells and normal physiology (PF-7006, PF-3837; Ki<0.5 nM; cellular IC50 2-6 nM). Treatment of tumor cells in vitro with PF-7006 modulates expected Mps1-dependent biology as demonstrated by molecular and phenotypic measures (reduced pHH3-Ser10 levels, shorter duration of mitosis, micro-nucleation, and apoptosis). Tumor-bearing mice treated with PF-7006 exhibit tumor growth inhibition concomitant with pharmacodynamic modulation of a downstream biomarker (pHH3-Ser10). Unfortunately, efficacy only occurs at drug exposures that cause dose-limiting body weight loss, gastrointestinal toxicities, and neutropenia. Mps1 inhibitor toxicities may be mitigated by inducing G1 cell cycle arrest in Rb1-competent cells with the cyclin-dependent kinase-4/6 inhibitor palbociclib. Using an isogenic cellular model system, PF-7006 is shown to be selectively cytotoxic to Rb1-deficient cells relative to Rb1-competent cells (also a measure of kinase selectivity). Human bone marrow cells pretreated with palbociclib have decreased PF-7006-dependent apoptosis relative to cells without palbociclib pretreatment. Collectively, this study raises a concern that single agent therapies inhibiting Mps1 will not be well-tolerated clinically but may be when combined with a selective CDK4/6 drug.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Histones/metabolism , Humans , Intestine, Small/pathology , Mice , Mice, SCID , Mitosis/drug effects , Phosphorylation , Piperazines/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/toxicity , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Transplantation, Heterologous
2.
J Ocul Pharmacol Ther ; 30(5): 429-39, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24720348

ABSTRACT

PURPOSE: Taprenepag isopropyl is an EP2 receptor agonist that is in development for the treatment of glaucoma. Iritis, photophobia, and increased corneal thickness observed in a Phase 2 clinical trial with taprenepag isopropyl were not previously observed in topical ocular toxicity studies in rabbits and dogs. In vivo studies using cynomolgus monkeys and in vitro models were used to elucidate the mechanisms underlying these ocular events. METHODS: Monkeys were dosed daily for 28 days in 1 eye with taprenepag and in the other with vehicle control. Complete ophthalmic examinations were performed at baseline and weekly thereafter. Serial sections of eyes were examined histopathologically at the end of the study. Recovery after the discontinuation of taprenepag was assessed for 28 days in the monkeys in the high-dose group. In vitro studies evaluated cell viability, paracellular permeability, and cytokine induction with human corneal epithelial or endothelial cell cultures. RESULTS: Monkeys demonstrated a dose-related incidence of iritis and increased corneal thickness that resolved within 28 days of discontinuing taprenepag. There was no evidence in vivo of taprenepag toxicity to the corneal endothelium or epithelium. Cell viability of stratified epithelial cells was primarily affected by excipients and was similar to Xalatan(®). The viability of HCEC-12 cells was not affected by taprenepag at concentrations up to 100 µM. CONCLUSIONS: The lack of in vivo or in vitro endothelial cytotoxicity and the reversibility of the increase in corneal thickness and iritis in the monkey provide confidence to permit further clinical development of taprenepag.


Subject(s)
Acetates/administration & dosage , Drug Discovery/trends , Epithelium, Corneal/drug effects , Epithelium, Corneal/pathology , Glaucoma/drug therapy , Receptors, Prostaglandin E, EP2 Subtype/agonists , Sulfonamides/administration & dosage , Administration, Ophthalmic , Administration, Topical , Animals , Cattle , Cells, Cultured , Dogs , Drug Discovery/methods , Glaucoma/pathology , Humans , Macaca fascicularis , Male , Treatment Outcome
3.
Toxicol Sci ; 135(2): 451-64, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23872713

ABSTRACT

The fibroblast growth factor receptors (FGFR) play a major role in angiogenesis and are desirable targets for the development of therapeutics. Groups of Wistar Han rats were dosed orally once daily for 4 days with a small molecule pan-FGFR inhibitor (5mg/kg) or once daily for 6 days with a small molecule MEK inhibitor (3mg/kg). Serum phosphorous and FGF23 levels increased in all rats during the course of the study. Histologically, rats dosed with either drug exhibited multifocal, multiorgan soft tissue mineralization. Expression levels of the sodium phosphate transporter Npt2a and the vitamin D-metabolizing enzymes Cyp24a1 and Cyp27b1 were modulated in kidneys of animals dosed with the pan-FGFR inhibitor. Both inhibitors decreased ERK phosphorylation in the kidneys and inhibited FGF23-induced ERK phosphorylation in vitro in a dose-dependent manner. A separate cardiovascular outcome study was performed to monitor hemodynamics and cardiac structure and function of telemetered rats dosed with either the pan-FGFR inhibitor or MEK inhibitor for 3 days. Both compounds increased blood pressure (~+ 17 mmHg), decreased heart rate (~-75 bpm), and modulated echocardiography parameters. Our data suggest that inhibition of FGFR signaling following administration of either pan-FGFR inhibitor or MEK inhibitor interferes with the FGF23 pathway, predisposing animals to hyperphosphatemia and a tumoral calcinosis-like syndrome in rodents.


Subject(s)
Cardiovascular System/physiopathology , Fibroblast Growth Factors/metabolism , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction , Animals , Base Sequence , Cardiovascular System/metabolism , Cell Line , DNA Primers , Fibroblast Growth Factor-23 , Humans , Phosphorylation , Rats , Rats, Wistar
4.
Toxicol In Vitro ; 25(5): 1061-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21513789

ABSTRACT

Thiabendazole (TBZ), an antihelminthic and antifungal agent, is associated with a host of adverse effects including nephrotoxicity, hepatotoxicity, and teratogenicity. Bioactivation of the primary metabolite of TBZ, 5-hydroxythiabendazole, has been proposed to yield a reactive intermediate. Here we show that this reactive intermediate can be catalyzed by myeloperoxidase (MPO), a neutrophil-bourne peroxidase. Using a cell viability endpoint, we examined the toxicity of TBZ, 5OH-TBZ, and MPO-generated metabolites in cell-based models including primary rat proximal tubule epithelial cells, NRK-52E rat proximal tubule cells, and H9C2 rat myocardial cells. Timecourse experiments with MPO showed complete turnover of 5OH-TBZ within 15 min and a dramatic leftward shift in dose-response curves after 12h. After a 24h exposure in vitro, the LC(50) of this reactive intermediate was 23.3 ± 0.2 µM reduced from greater than 200 µM from 5OH-TBZ alone, an approximately 10-fold decrease. LC(50) values were equal in all cell types used. Comparison of lactate dehydrogenase leakage and caspase 3/7 activity revealed that cell death caused by the reactive intermediate is primarily associated with necrosis rather than apoptosis. This toxicity can be completely rescued via incubation with rutin, an inhibitor of MPO. These results suggest that MPO-mediated biotransformation of 5OH-TBZ yields a reactive intermediate which may play a role in TBZ-induced toxicity.


Subject(s)
Antifungal Agents/toxicity , Neutrophils/pathology , Peroxidase/metabolism , Thiabendazole/analogs & derivatives , Animals , Biotransformation , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/pathology , L-Lactate Dehydrogenase/metabolism , Male , Necrosis , Neutrophils/metabolism , Rats , Rats, Wistar , Rutin/pharmacology , Thiabendazole/toxicity
5.
Toxicol In Vitro ; 24(4): 1324-31, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20144907

ABSTRACT

Preservatives are used in ocular medications to prevent microbial contamination. The use of benzalkonium chloride (BAC), the most widely used preservative in ocular medications, has been scrutinized with a number of studies indicating its toxicity to monolayer cultures of corneal and conjunctival epithelial cells. The purpose of this study was to evaluate and compare the toxicity of BAC and other preservatives and common components of ocular formulations on monolayer and stratified air-lifted cultures of Chang conjunctival cells. Air-lifting Chang cells grown on transwell filters increased stratification as assessed by transepithelial electrical resistance and histology. Unlike monolayer cultures in which ocular medications containing BAC caused near complete loss of cell viability, stratified, air-lifted cultures were not affected by the presence of BAC in ocular medications with up to 30-min exposures. Stratification shifted the dose-response curve to the right for benzalkonium chloride, thimerosal, chlorhexidine digluconate, potassium sorbate and EDTA. These results demonstrate that stratification significantly affects cell viability of Chang conjunctival cells in response to preservatives and additives of ophthalmic preparations.


Subject(s)
Benzalkonium Compounds/toxicity , Conjunctiva/drug effects , Preservatives, Pharmaceutical/toxicity , Cell Line , Cell Survival/drug effects , Chlorhexidine/analogs & derivatives , Chlorhexidine/toxicity , Conjunctiva/cytology , Dose-Response Relationship, Drug , Edetic Acid/toxicity , Ophthalmic Solutions , Sorbic Acid/toxicity , Thimerosal/toxicity , Toxicity Tests
6.
Breast Cancer Res ; 8(2): R18, 2006.
Article in English | MEDLINE | ID: mdl-16584539

ABSTRACT

INTRODUCTION: Activation of the type I insulin-like growth factor receptor (IGFIR) promotes proliferation and inhibits apoptosis in a variety of cell types. Transgenic mice expressing a constitutively active IGFIR or IGF-I develop mammary tumors and increased levels of IGFIR have been detected in primary breast cancers. However, the contribution of IGFIR activation in promoting breast cancer progression remains unknown. Mammary epithelial cell lines grown in three-dimensional cultures form acinar structures that mimic the round, polarized, hollow and growth-arrested features of mammary alveoli. We used this system to determine how proliferation and survival signaling by IGFIR activation affects breast epithelial cell biology and contributes to breast cancer progression. METHODS: Pooled, stable MCF-10A breast epithelial cells expressing wild-type IGFIR or kinase-dead IGFIR (K1003A) were generated using retroviral-mediated gene transfer. The effects of over-expression of wild-type or kinase-dead IGFIR on breast epithelial cell biology were analyzed by confocal microscopy of three-dimensional cultures. The contribution of signaling pathways downstream of IGFIR activation to proliferation and apoptosis were determined by pharmacological inhibition of phosphatidylinositol 3' kinase (PI3K) with LY294002, MAP kinase kinase (MEK) with UO126 and mammalian target of rapamycin (mTOR) with rapamycin. RESULTS: We found that MCF-10A cells over-expressing the IGFIR formed large, misshapen acinar structures with filled lumina and disrupted apico-basal polarization. This phenotype was ligand-dependent, occurring with IGF-I or supraphysiological doses of insulin, and did not occur in cells over-expressing the kinase-dead receptor. We observed increased proliferation, decreased apoptosis and increased phosphorylation of Ser473 of Akt and Ser2448 of mTOR throughout IGFIR structures. Inhibition of PI3K with LY294002 or MEK with UO126 prevented the development of acinar structures from IGFIR-expressing but not control cells. The mTOR inhibitor rapamycin failed to prevent IGFIR-induced hyperproliferation and survival signaling. CONCLUSION: Increased proliferation and survival signaling as well as loss of apico-basal polarity by IGFIR activation in mammary epithelial cells may promote early lesions of breast cancer. Three-dimensional cultures of MCF-10A cells over-expressing the IGFIR are a useful model with which to study the role of IGFIR signaling in breast cancer progression and for characterizing the effects of chemotherapeutics targeted to IGFIR signaling.


Subject(s)
Apoptosis/physiology , Breast Neoplasms/pathology , Epithelial Cells/cytology , Epithelial Cells/physiology , Receptor, IGF Type 1/genetics , Signal Transduction/physiology , Breast Neoplasms/physiopathology , Cell Division/drug effects , Cell Line, Tumor , Cell Polarity , Cell Survival , Cells, Cultured , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Gene Transfer Techniques , Humans , Insulin/pharmacology , Morphogenesis , Retroviridae , Signal Transduction/drug effects
7.
Biophys J ; 86(3): 1470-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990474

ABSTRACT

Drosophila Big Brain (BIB) is a transmembrane protein encoded by the neurogenic gene big brain (bib), which is important for early development of the fly nervous system. BIB expressed in Xenopus oocytes is a monovalent cation channel modulated by tyrosine kinase signaling. Results here demonstrate that the BIB conductance shows voltage- and dose-dependent block by extracellular divalent cations Ca(2+) and Ba(2+) but not by Mg(2+) in wild-type channels. Site-directed mutagenesis of negatively charged glutamate (Glu(274)) and aspartate (Asp(253)) residues had no effect on divalent cation block. However, mutation of a conserved glutamate at position 71 (Glu(71)) in the first transmembrane domain (M1) altered channel properties. Mutation of Glu(71) to Asp introduced a new sensitivity to block by extracellular Mg(2+); substitutions with asparagine or glutamine decreased whole-cell conductance; and substitution with lysine compromised plasma membrane expression. Block by divalent cations is important in other ion channels for voltage-dependent function, enhanced signal resolution, and feedback regulation. Our data show that the wild-type BIB conductance is attenuated by external Ca(2+), suggesting that endogenous divalent cation block might be relevant for enhancing signal resolution or voltage dependence for the native signaling process in neuronal cell fate determination.


Subject(s)
Barium/pharmacology , Calcium/pharmacology , Drosophila Proteins/physiology , Ion Channel Gating/physiology , Ion Channels/physiology , Magnesium/pharmacology , Membrane Proteins/physiology , Oocytes/physiology , Animals , Cations, Divalent/pharmacology , Cells, Cultured , Drosophila , Drosophila Proteins/drug effects , Extracellular Fluid/metabolism , Ion Channel Gating/drug effects , Ion Channels/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/drug effects , Mutagenesis, Site-Directed , Oocytes/drug effects , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Structure-Activity Relationship , Xenopus laevis
8.
J Neurosci ; 22(7): 2530-40, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11923418

ABSTRACT

Big brain (bib) is a neurogenic gene that when mutated causes defects in cell fate determination during Drosophila neurogenesis through an unknown mechanism. The protein Big Brain (BIB) has sequence identity with the major intrinsic protein family that includes the water- and ion-conducting aquaporin channels. We show here that BIB expressed heterologously in Xenopus oocytes provides a voltage-insensitive, nonselective cation channel function with permeability to K+ > Na+ >> tetraethylammonium. The conductance, activated in response to endogenous signaling pathways in BIB-expressing oocytes, is decreased after treatment with 20 microm insulin and is enhanced with 10 microm lavendustin A, a tyrosine kinase inhibitor. Western blot analysis confirms that BIB is tyrosine-phosphorylated. Both tyrosine phosphorylation and the potentiating effect of lavendustin A are removed by partial deletion of the C terminus (amino acids 317-700). Current activation is not observed in control oocytes or in oocytes expressing a nonfunctional mutant (BIB E71N) that appears to be expressed on the plasma membrane by confocal microscopy and Western blotting. These results indicate that BIB can participate in tyrosine kinase-regulated transmembrane signaling and may suggest a role for membrane depolarization in the neurogenic function of BIB in early development.


Subject(s)
Cations/metabolism , Drosophila Proteins , Ion Channels/metabolism , Membrane Proteins/metabolism , Oocytes/metabolism , Animals , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Dose-Response Relationship, Drug , Drosophila , Enzyme Inhibitors/pharmacology , Gene Expression , Insulin/pharmacology , Ion Channels/drug effects , Ion Transport/drug effects , Ion Transport/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/genetics , Microinjections , Mutagenesis, Site-Directed , Oocytes/cytology , Oocytes/drug effects , Osmolar Concentration , Patch-Clamp Techniques , Phosphorylation , RNA, Complementary/administration & dosage , Signal Transduction/physiology , Structure-Activity Relationship , Transfection , Water/metabolism , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...