Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nanomedicine (Lond) ; 10(19): 2989-3001, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26419658

ABSTRACT

AIMS: RALA is a novel 30 mer bioinspired amphipathic peptide that is showing promise for gene delivery. Here, we used RALA to deliver the FK506-binding protein like - FKBPL gene (pFKBPL) - a novel member of the immunophilin protein family. FKBPL is a secreted protein, with overexpression shown to inhibit angiogenesis, tumor growth and stemness, through a variety of intra- and extracellular signaling mechanisms. We also elucidated proangiogenic activity and stemness after utilizing RALA to deliver siRNA (siFKBPL). MATERIALS & METHODS: The RALA/pFKBPL and RALA/siFKBPL nanoparticles were characterized in terms of size, charge, stability and toxicity. Overexpression and knockdown of FKBPL was assessed in vitro and in vivo. RESULTS: RALA delivered both pFKBPL and siFKBPL with less cytotoxicity than commercially available counterparts. In vivo, RALA/pFKBPL delivery retarded tumor growth, and prolonged survival with an associated decrease in angiogenesis, while RALA/siFKBPL had no effect on tumor growth rate or survival, but resulted in an increase in angiogenesis and stemness. CONCLUSION: RALA is an effective delivery system for both FKBPL DNA and RNAi and highlights an alternative therapeutic approach to harnessing FKBPL's antiangiogenic and antistemness activity.

2.
Oncotarget ; 6(14): 12209-23, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25906750

ABSTRACT

FK506-binding protein-like (FKBPL) has established roles as an anti-tumor protein, with a therapeutic peptide based on this protein, ALM201, shortly entering phase I/II clinical trials. Here, we evaluated FKBPL's prognostic ability in primary breast cancer tissue, represented on tissue microarrays (TMA) from 3277 women recruited into five independent retrospective studies, using immunohistochemistry (IHC). In a meta-analysis, FKBPL levels were a significant predictor of BCSS; low FKBPL levels indicated poorer breast cancer specific survival (BCSS) (hazard ratio (HR) = 1.30, 95% confidence interval (CI) 1.14-1.49, p < 0.001). The prognostic impact of FKBPL remained significant after adjusting for other known prognostic factors (HR = 1.25, 95% CI 1.07-1.45, p = 0.004). For the sub-groups of 2365 estrogen receptor (ER) positive patients and 1649 tamoxifen treated patients, FKBPL was significantly associated with BCSS (HR = 1.34, 95% CI 1.13-1.58, p < 0.001, and HR = 1.25, 95% CI 1.04-1.49, p = 0.02, respectively). A univariate analysis revealed that FKBPL was also a significant predictor of relapse free interval (RFI) within the ER positive patient group, but it was only borderline significant within the smaller tamoxifen treated patient group (HR = 1.32 95% CI 1.05-1.65, p = 0.02 and HR = 1.23 95% CI 0.99-1.54, p = 0.06, respectively). The data suggests a role for FKBPL as a prognostic factor for BCSS, with the potential to be routinely evaluated within the clinic.


Subject(s)
Breast Neoplasms/genetics , Immunophilins/genetics , Immunophilins/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cohort Studies , Female , Humans , Precision Medicine , Prognosis , Survival Analysis , Tacrolimus Binding Proteins
3.
Arterioscler Thromb Vasc Biol ; 35(4): 845-54, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25767277

ABSTRACT

OBJECTIVE: The antitumor effects of FK506-binding protein like (FKBPL) and its extracellular role in angiogenesis are well characterized; however, its role in physiological/developmental angiogenesis and the effect of FKBPL ablation has not been evaluated. This is important as effects of some angiogenic proteins are dosage dependent. Here we evaluate the regulation of FKBPL secretion under angiogenic stimuli, as well as the effect of FKBPL ablation in angiogenesis using mouse and zebrafish models. APPROACH AND RESULTS: FKBPL is secreted maximally by human microvascular endothelial cells and fibroblasts, and this was specifically downregulated by proangiogenic hypoxic signals, but not by the angiogenic cytokines, VEGF or IL8. FKBPL's critical role in angiogenesis was supported by our inability to generate an Fkbpl knockout mouse, with embryonic lethality occurring before E8.5. However, whilst Fkbpl heterozygotic embryos showed some vasculature irregularities, the mice developed normally. In murine angiogenesis models, including the ex vivo aortic ring assay, in vivo sponge assay, and tumor growth assay, Fkbpl(+/-) mice exhibited increased sprouting, enhanced vessel recruitment, and faster tumor growth, respectively, supporting the antiangiogenic function of FKBPL. In zebrafish, knockdown of zFkbpl using morpholinos disrupted the vasculature, and the phenotype was rescued with hFKBPL. Interestingly, this vessel disruption was ineffective when zcd44 was knocked-down, supporting the dependency of zFkbpl on zCd44 in zebrafish. CONCLUSIONS: FKBPL is an important regulator of angiogenesis, having an essential role in murine and zebrafish blood vessel development. Mouse models of angiogenesis demonstrated a proangiogenic phenotype in Fkbpl heterozygotes.


Subject(s)
Aorta/metabolism , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Immunophilins/metabolism , Neovascularization, Pathologic , Tacrolimus Binding Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Carcinoma, Lewis Lung/pathology , Cell Hypoxia , Female , Gene Expression Regulation, Developmental , Genotype , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunophilins/genetics , MCF-7 Cells , Male , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Physiologic , Phenotype , Signal Transduction , Tacrolimus Binding Proteins/genetics , Time Factors , Tumor Burden , Zebrafish , Zebrafish Proteins/genetics
4.
Clin Cancer Res ; 19(14): 3881-93, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23741069

ABSTRACT

PURPOSE: FK506-binding protein like (FKBPL) and its peptide derivative, AD-01, have already shown tumor growth inhibition and CD44-dependent antiangiogenic activity. Here, we explore the ability of AD-01 to target CD44-positive breast cancer stem cells (BCSC). EXPERIMENTAL DESIGN: Mammosphere assays and flow cytometry were used to analyze the effect of FKBPL overexpression/knockdown and AD-01 treatment ± other anticancer agents on BCSCs using breast cancer cell lines (MCF-7/MDA-231/ZR-75), primary patient samples, and xenografts. Delays in tumor initiation were evaluated in vivo. The anti-stem cell mechanisms were determined using clonogenic assays, quantitative PCR (qPCR), and immunofluorescence. RESULTS: AD-01 treatment was highly effective at inhibiting the BCSC population by reducing mammosphere-forming efficiency and ESA(+)/CD44(+)/CD24(-) or aldehyde dehydrogenase (ALDH)(+) cell subpopulations in vitro and tumor initiation in vivo. The ability of AD-01 to inhibit the self-renewal capacity of BCSCs was confirmed; mammospheres were completely eradicated by the third generation. The mechanism seems to be due to AD-01-mediated BCSC differentiation shown by a significant decrease in the number of holoclones and an associated increase in meroclones/paraclones; the stem cell markers, Nanog, Oct4, and Sox2, were also significantly reduced. Furthermore, we showed additive inhibitory effects when AD-01 was combined with the Notch inhibitor, DAPT. AD-01 was also able to abrogate a chemo- and radiotherapy-induced enrichment in BCSCs. Finally, FKBPL knockdown led to an increase in Nanog/Oct4/Sox2 and an increase in BCSCs, highlighting a role for endogenous FKBPL in stem cell signaling. CONCLUSIONS: AD-01 has dual antiangiogenic and anti-BCSC activity, which will be advantageous as this agent enters clinical trial.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Hyaluronan Receptors/metabolism , Immunophilins/pharmacology , Neoplastic Stem Cells/drug effects , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Drug Resistance, Neoplasm , Female , Humans , Immunophilins/metabolism , MCF-7 Cells , Mice , Mice, SCID , Radiation Tolerance , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Signal Transduction , Spheroids, Cellular/drug effects , Tacrolimus Binding Proteins , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
PLoS One ; 8(2): e55075, 2013.
Article in English | MEDLINE | ID: mdl-23457460

ABSTRACT

FK506 binding protein-like (FKBPL) and its peptide derivatives exert potent anti-angiogenic activity in vitro and in vivo and control tumour growth in xenograft models, when administered exogenously. However, the role of endogenous FKBPL in angiogenesis is not well characterised. Here we investigated the molecular effects of the endogenous protein and its peptide derivative, AD-01, leading to their anti-migratory activity. Inhibition of secreted FKBPL using a blocking antibody or siRNA-mediated knockdown of FKBPL accelerated the migration of human microvascular endothelial cells (HMEC-1). Furthermore, MDA-MB-231 tumour cells stably overexpressing FKBPL inhibited tumour vascular development in vivo suggesting that FKBPL secreted from tumour cells could inhibit angiogenesis. Whilst FKBPL and AD-01 target CD44, the nature of this interaction is not known and here we have further interrogated this aspect. We have demonstrated that FKBPL and AD-01 bind to the CD44 receptor and inhibit tumour cell migration in a CD44 dependant manner; CD44 knockdown abrogated AD-01 binding as well as its anti-migratory activity. Interestingly, FKBPL overexpression and knockdown or treatment with AD-01, regulated CD44 expression, suggesting a co-regulatory pathway for these two proteins. Downstream of CD44, alterations in the actin cytoskeleton, indicated by intense cortical actin staining and a lack of cell spreading and communication were observed following treatment with AD-01, explaining the anti-migratory phenotype. Concomitantly, AD-01 inhibited Rac-1 activity, up-regulated RhoA and the actin binding proteins, profilin and vinculin. Thus the anti-angiogenic protein, FKBPL, and AD-01, offer a promising and alternative approach for targeting both CD44 positive tumours and vasculature networks.


Subject(s)
Cell Movement/drug effects , Cytoskeleton/metabolism , Hyaluronan Receptors/metabolism , Immunophilins/metabolism , Peptides/chemistry , Peptides/pharmacology , Actins/metabolism , Amino Acid Sequence , Cell Line , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Hyaluronan Receptors/genetics , Immunophilins/analysis , Molecular Sequence Data , Peptides/chemical synthesis , Signal Transduction/drug effects , Tacrolimus Binding Proteins , Tubulin/metabolism , rho-Associated Kinases/metabolism
6.
Clin Cancer Res ; 17(5): 1044-56, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21364036

ABSTRACT

PURPOSE: Antiangiogenic therapies can be an important adjunct to the management of many malignancies. Here we investigated a novel protein, FKBPL, and peptide derivative for their antiangiogenic activity and mechanism of action. EXPERIMENTAL DESIGN: Recombinant FKBPL (rFKBPL) and its peptide derivative were assessed in a range of human microvascular endothelial cell (HMEC-1) assays in vitro. Their ability to inhibit proliferation, migration, and Matrigel-dependent tubule formation was determined. They were further evaluated in an ex vivo rat model of neovascularization and in two in vivo mouse models of angiogenesis, that is, the sponge implantation and the intravital microscopy models. Antitumor efficacy was determined in two human tumor xenograft models grown in severe compromised immunodeficient (SCID) mice. Finally, the dependence of peptide on CD44 was determined using a CD44-targeted siRNA approach or in cell lines of differing CD44 status. RESULTS: rFKBPL inhibited endothelial cell migration, tubule formation, and microvessel formation in vitro and in vivo. The region responsible for FKBPL's antiangiogenic activity was identified, and a 24-amino acid peptide (AD-01) spanning this sequence was synthesized. It was potently antiangiogenic and inhibited growth in two human tumor xenograft models (DU145 and MDA-231) when administered systemically, either on its own or in combination with docetaxel. The antiangiogenic activity of FKBPL and AD-01 was dependent on the cell-surface receptor CD44, and signaling downstream of this receptor promoted an antimigratory phenotype. CONCLUSION: FKBPL and its peptide derivative AD-01 have potent antiangiogenic activity. Thus, these agents offer the potential of an attractive new approach to antiangiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Immunophilins/therapeutic use , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Animals , Blotting, Western , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Docetaxel , Endothelial Cells/drug effects , Hyaluronan Receptors/genetics , Immunophilins/chemistry , Immunophilins/pharmacology , Immunoprecipitation , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasms/drug therapy , Peptide Fragments/therapeutic use , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Tacrolimus Binding Proteins , Taxoids/pharmacology , Taxoids/therapeutic use , Xenograft Model Antitumor Assays
7.
Biochem Soc Trans ; 39(2): 663-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21428958

ABSTRACT

FKBPs (FK506-binding proteins) have long been recognized as key regulators of the response to immunosuppressant drugs and as co-chaperones of steroid receptor complexes. More recently, evidence has emerged suggesting that this diverse protein family may also represent cancer biomarkers owing to their roles in cancer progression and response to treatment. FKBPL (FKBP-like) is a novel FKBP with roles in GR (glucocorticoid receptor), AR (androgen receptor) and ER (oestrogen receptor) signalling. FKBPL binds Hsp90 (heat-shock protein 90) and modulates translocation, transcriptional activation and phosphorylation of these steroid receptors. It has been proposed as a novel prognostic and predictive biomarker, where high levels predict for increased recurrence-free survival in breast cancer patients and enhanced sensitivity to endocrine therapy. Since this protein family has roles in a plethora of signalling pathways, its members represent novel prognostic markers and therapeutic targets for cancer diagnosis and treatment.


Subject(s)
Biomarkers, Tumor/physiology , Immunophilins/physiology , Neoplasms/diagnosis , Tacrolimus Binding Proteins/physiology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Immunophilins/genetics , Immunophilins/metabolism , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction/physiology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
8.
Cancer Res ; 70(3): 1090-100, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20103631

ABSTRACT

The HSP90 chaperone and immunophilin FKBPL is an estrogen-responsive gene that interacts with estogen receptor alpha (ERalpha) and regulates its levels. In this study, we explored the effects of FKBPL on breast cancer proliferation. Breast cancer cells stably overexpressing FKBPL became dependent on estrogen for their growth and were dramatically more sensitive to the antiestrogens tamoxifen and fulvestrant, whereas FKBPL knockdown reverses this phenotype. FKBPL knockdown also decreased the levels of the cell cycle inhibitor p21WAF1 and increased ERalpha phosphorylation on Ser(118) in response to 17beta-estradiol and tamoxifen. In support of the likelihood that these effects explained FKBPL-mediated cell growth inhibition and sensitivity to endocrine therapies, FKBPL expression was correlated with increased overall survival and distant metastasis-free survival in breast cancer patients. Our findings suggest that FKBPL may have prognostic value based on its impact on tumor proliferative capacity and sensitivity to endocrine therapies, which improve outcome.


Subject(s)
Estrogen Receptor alpha/metabolism , Immunophilins/physiology , Signal Transduction/physiology , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogens/pharmacology , Fulvestrant , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunophilins/genetics , Immunophilins/metabolism , Immunoprecipitation , Kaplan-Meier Estimate , Meta-Analysis as Topic , Phosphorylation/drug effects , Protein Binding , RNA Interference , Serine/metabolism , Signal Transduction/genetics , Tacrolimus Binding Proteins , Transfection
9.
Mol Carcinog ; 48(2): 110-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18618592

ABSTRACT

Cytochrome P450 1B1 (CYP1B1) mRNA is constitutively expressed in most normal extra-hepatic tissues; however the protein is not detectable in these tissues but is expressed in a wide variety of tumors. CYP1B1 is responsible for the activation of a number of carcinogens present in tobacco smoke and food. A surgical model of rat esophageal tumorigenesis, promoted by gastric or duodenal reflux was used to determine CYP1B1 expression in premalignant esophageal tissue. Immunohistochemistry was performed using a modified amplified fluorescein tyramide protocol. CYP1B1 was not observed in normal esophageal mucosa, submucosa, or muscularis mucosa. Animals exposed to gastric reflux developed mild hyperplasia. Varying degrees of hyperplasia were observed in the duodenal reflux group. All regions of hyperplasia showed moderate or strong CYP1B1 immunoreactivity. Duodenal reflux induced a small number of premalignant changes: immunoreactivity was absent from the epithelium of squamous dysplasia (0/10), Barrett's esophagus (0/7), and majority of dysplastic Barrett's esophagus (1/4). Moderate or strong immunoreactivity was observed in the majority (7/8) of squamous cell carcinomas (SCCs) in situ. Immunoreactivity was also observed in the lamina propria and submucosa in association with inflammation, regardless of the severity of inflammation. The expression of CYP1B1 in hyperplasia, SCCs in situ, or in association with inflammation may increase the production of carcinogenic metabolites, which may promote esophageal tumorigenesis.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Duodenogastric Reflux/complications , Esophageal Neoplasms/enzymology , Gastroesophageal Reflux/complications , Animals , Antibody Specificity , Aryl Hydrocarbon Hydroxylases/immunology , Blotting, Western , Cytochrome P-450 CYP1B1 , Duodenogastric Reflux/enzymology , Esophageal Neoplasms/etiology , Female , Gastroesophageal Reflux/enzymology , Immunohistochemistry , Mice , Mice, Knockout , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
10.
Endocrinology ; 149(11): 5724-34, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18669603

ABSTRACT

FKBP-like (FKBPL) protein is a novel immunophilin-like protein that plays a role in the cellular stress response. Its three tetratricopeptide repeat motifs are homologous to the heat shock protein 90 interaction sites of other immunophilins that have roles in steroid hormone receptor signaling. In this study, using biomolecular complementation and coimmunoprecipitation techniques, we show that FKBPL also colocalizes and interacts with the components of the heat shock protein 90-glucocorticoid receptor (GR) complex and demonstrate that the PPIase domain of FKBPL is important for the interaction between this complex and the dynein motor protein, dynamitin. Treatment of DU145 cells with the GR ligand, dexamethasone, induced a rapid and coordinated translocation of both GR and FKBPL to the nucleus; this response was perturbed when FKBPL was knocked down with a targeted small interfering RNA. Furthermore, overexpression of FKBPL increased GR protein levels and transactivation of a luciferase reporter gene in response to dexamethasone in DU145 cells. However, these responses were cell line dependent. In summary, these data suggest that FKBPL can be classed as a new member of the FKBP protein family with a role in steroid receptor complexes and signaling.


Subject(s)
Immunophilins/metabolism , Immunophilins/physiology , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/physiology , Cell Nucleus/metabolism , Humans , Immunophilins/antagonists & inhibitors , Immunophilins/genetics , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , RNA, Small Interfering/pharmacology , Receptors, Steroid/metabolism , Signal Transduction/physiology , Tacrolimus Binding Proteins , Transcriptional Activation/drug effects , Transfection , Tumor Cells, Cultured
11.
Carcinogenesis ; 27(12): 2519-27, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16777992

ABSTRACT

BACKGROUND: Familial breast cancer is associated with mutations in several genes (BRCA1, BRCA2, p53, ATM) whose protein products protect against radiation-induced genotoxicity. This study tested whether sporadic breast cancer was associated with constitutive radiation hypersensitivity. METHODS: Blood lymphocytes and EBV-transformed lymphoblasts from patients with newly diagnosed breast cancer and controls without cancer were evaluated for ionizing radiation (IR)-induced chromosomal aberrations and cell cycle delays. Lymphoblasts from patients with ataxia telangiectasia (AT) and heterozygous AT carriers were tested as positive controls for radiation hypersensitivity. RESULTS: Lymphoblasts from AT patients and AT carriers displayed G2-irradiation, chromosomal hypersensitivity (GICH). Irradiated G2 phase lymphocytes from breast cancer cases and controls displayed 3-fold inter-individual variation in frequencies of chromatid damage. However, the percentage of breast cancer cases with damage frequencies in excess of 2 SD of the control mean (8/102 or 8%) was not significantly elevated compared to controls (2/48 or 4%, P=0.5). Lymphoblasts sampled 24 h after 3 Gy of IR also varied in the ratios of cells with 4N and 2N DNA content (4N/2N ratio), as a measure of cell cycle checkpoint function. 4N/2N ratios in irradiated lymphoblasts were strongly correlated with the fractions of S phase cells in un-irradiated control cultures (Pearson's correlation coefficient, r=0.87). After normalization to S fraction, the radiation-induced increment in the 4N/2N ratio was significantly elevated in AT lymphoblasts but not in lymphoblasts from AT carriers. The fraction of breast cancer cases with reduced checkpoint function (2/45 or 4%) was equal to the control fraction (2/45 or 4%). For breast cancer cases and controls, GICH in primary lymphocytes was not associated with reduced cell cycle checkpoint function in lymphoblasts. CONCLUSION: Constitutive radiation hypersensitivity in blood lymphocytes and lymphoblasts was not a useful biomarker for identifying women at increased risk of breast cancer.


Subject(s)
Breast Neoplasms/epidemiology , Cell Cycle/genetics , Cell Cycle/radiation effects , Mutation , Ataxia Telangiectasia Mutated Proteins , Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Genes, p53 , Humans , Middle Aged , Protein Serine-Threonine Kinases/genetics , Racial Groups , Risk Factors , Smoking , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...