Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 8(6): e66343, 2013.
Article in English | MEDLINE | ID: mdl-23822995

ABSTRACT

Parathyroid hormone-related protein (PTHrP) is a key component in breast development and breast tumour biology. PTHrP has been discovered as a causative agent of hypercalcaemia of malignancy and is also one of the main factors implicated in breast cancer mediated osteolysis. Clinical studies have determined that PTHrP expression by primary breast cancers was an independent predictor of improved prognosis. Furthermore, PTHrP has been demonstrated to cause tumour cell death both in vitro and in vivo. Apo2L/TRAIL is a promising new anti-cancer agent, due to its ability to selectively induce apoptosis in cancer cells whilst sparing most normal cells. However, some cancer cells are resistant to Apo2L/TRAIL-induced apoptosis thus limiting its therapeutic efficacy. The effects of PTHrP on cell death signalling pathways initiated by Apo2L/TRAIL were investigated in breast cancer cells. Expression of PTHrP in Apo2L/TRAIL resistant cell line MCF-7 sensitised these cells to Apo2L/TRAIL-induced apoptosis. The actions of PTHrP resulted from intracellular effects, since exogenous treatment of PTHrP had no effect on Apo2L/TRAIL-induced apoptosis. Apo2L/TRAIL-induced apoptosis in PTHrP expressing cells occurred through the activation of caspase-10 resulting in caspase-9 activation and induction of apoptosis through the effector caspases, caspase-6 and -7. PTHrP increased cell surface expression of Apo2L/TRAIL death receptors, TRAIL-R1 and TRAIL-R2. Antagonistic antibodies against the death receptors demonstrated that Apo2L/TRAIL mediated its apoptotic signals through activation of the TRAIL-R2 in PTHrP expressing breast cancer cells. These studies reveal a novel role for PTHrP with Apo2L/TRAIL that maybe important for future diagnosis and treatment of breast cancer.


Subject(s)
APOBEC Deaminases/metabolism , Breast Neoplasms/pathology , Muscle Proteins/metabolism , Parathyroid Hormone-Related Protein/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Humans
2.
Int J Oncol ; 31(6): 1391-402, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982666

ABSTRACT

We investigated the effect of a novel histone deacetylase inhibitor, A-423378.0, on the colon carcinoma cell line HCT116 and genetically modified derivatives lacking either p21(cip1/waf1) or p53. HCT116 cell lines were incubated with A-423378.0 at different concentrations for 3-120 h. Cell viability, proliferation and apoptosis rates were determined and verified by western blot, detection of mitochondrial membrane potential breakdown DeltaPsi(m), activation of caspases-3, -8 and cytokeratin 18 cleavage. A subcutaneous xenograft model was established in NMRI mice with daily intraperitoneal injections of 10 mg/kg for 14 days. All three HCT116 cell lines responded to A-423378.0 treatment in a dose- and time-dependent manner via induction of apoptosis as measured by breakdown of DeltaPsi(m) and BrdU incorporation. We identified that A-423378.0 induced the expression of TRAIL and TRAIL receptor, especially TRAIL-R2/hDR5, which was up-regulated in HCT116 cells after treatment with A-423378.0. In vivo, a growth inhibitory effect was observed with HDAC-I treatment, which was paralleled by a down-regulation of PCNA and a concomitant induction of apoptosis. Treatment of wild-type or knock-out HCT116 cells with A-423378.0 exerts potent anti-proliferative and pro-apoptotic effects in vitro and in vivo. A-423378.0 was able to induce apoptosis in both p21(WAF1) and p53 deficient tumour cells, which appeared to be mediated by the intrinsic cell death pathway. Interestingly, the effects of A-423378.0 on the extrinsic cell death pathway through activation of TRAIL and its signalling pathway indicate that A-423378.0 may be a potent new therapeutic compound for the treatment of advanced colorectal cancer.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p21/physiology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Thiazoles/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Caspases/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Male , Membrane Potentials/drug effects , Mice , RNA, Messenger/analysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/analysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/analysis , bcl-2-Associated X Protein/physiology
3.
Cancer Res ; 66(10): 5363-70, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707463

ABSTRACT

Breast cancer is the most common carcinoma that metastasizes to bone. To examine the efficacy of recombinant soluble Apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) against breast cancer growth in bone, we established a mouse model in which MDA-MB-231 human breast cancer cells were transplanted directly into the marrow cavity of the tibiae of athymic nude mice producing osteolytic lesions in the area of injection. All vehicle-treated control animals developed large lesions that established in the marrow cavity, eroded the cortical bone, and invaded the surrounding soft tissue, as assessed by radiography, micro-computed tomography, and histology. In contrast, animals treated with recombinant soluble Apo2L/TRAIL showed significant conservation of the tibiae, with 85% reduction in osteolysis, 90% reduction in tumor burden, and no detectable soft tissue invasion. Tumor cells explanted from Apo2L/TRAIL-treated animals were significantly more resistant to the effects of Apo2L/TRAIL when compared with the cells explanted from the vehicle-treated control animals, suggesting that prolonged treatment with Apo2/TRAIL in vivo selects for a resistant phenotype. However, such resistance was readily reversed when Apo2L/TRAIL was used in combination with clinically relevant chemotherapeutic drugs, including taxol, etoposide, doxorubicin, cisplatin, or the histone deacetylase inhibitor suberoylanilide hydroxamic acid. These studies show for the first time that Apo2L/TRAIL can prevent breast cancer-induced bone destruction and highlight the potential of this ligand for the treatment of metastatic breast cancer in bone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Membrane Glycoproteins/pharmacology , Osteolysis/prevention & control , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/administration & dosage , Breast Neoplasms/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Green Fluorescent Proteins/biosynthesis , Humans , Membrane Glycoproteins/administration & dosage , Mice , Mice, Nude , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...