Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(11): 105311, 2023 11.
Article in English | MEDLINE | ID: mdl-37797694

ABSTRACT

While the role of endocytosis in focal adhesion turnover-coupled cell migration has been established in addition to its conventional role in cellular functions, the molecular regulators and precise molecular mechanisms that underlie this process remain largely unknown. In this study, we report that proto-oncoprotein hematopoietic PBX-interacting protein (HPIP) localizes to focal adhesions as well as endosomal compartments along with RUN FYVE domain-containing protein 3 (RUFY3) and Rab5, an early endosomal protein. HPIP contains two coiled-coil domains (CC1 and CC2) that are necessary for its association with Rab5 and RUFY3 as CC domain double mutant, that is, mtHPIPΔCC1-2 failed to support it. Furthermore, we show that HPIP and RUFY3 activate Rab5 by serving as noncanonical guanine nucleotide exchange factors of Rab5. In support of this, either deletion of coiled-coil domains or silencing of HPIP or RUFY3 impairs Rab5 activation and Rab5-dependent cell migration. Mechanistic studies further revealed that loss of HPIP or RUFY3 expression severely impairs Rab5-mediated focal adhesion disassembly, FAK activation, fibronectin-associated-ß1 integrin trafficking, and thus cell migration. Together, this study underscores the importance of HPIP and RUFY3 as noncanonical guanine nucleotide exchange factors of Rab5 and in integrin trafficking and focal adhesion turnover, which implicates in cell migration.


Subject(s)
Focal Adhesions , Guanine Nucleotide Exchange Factors , Cell Movement , Endocytosis , Focal Adhesions/genetics , Focal Adhesions/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism , Humans , Cell Line , Cell Line, Tumor
2.
Semin Cancer Biol ; 86(Pt 3): 1158-1174, 2022 11.
Article in English | MEDLINE | ID: mdl-36244530

ABSTRACT

Metabolic reprogramming is an important cancer hallmark that plays a key role in cancer malignancies and therapy resistance. Cancer cells reprogram the metabolic pathways to generate not only energy and building blocks but also produce numerous key signaling metabolites to impact signaling and epigenetic/transcriptional regulation for cancer cell proliferation and survival. A deeper understanding of the mechanisms by which metabolic reprogramming is regulated in cancer may provide potential new strategies for cancer targeting. Recent studies suggest that deregulated transcription factors have been observed in various human cancers and significantly impact metabolism and signaling in cancer. In this review, we highlight the key transcription factors that are involved in metabolic control, dissect the crosstalk between signaling and transcription factors in metabolic reprogramming, and offer therapeutic strategies targeting deregulated transcription factors for cancer treatment.


Subject(s)
Neoplasms , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Neoplasms/pathology , Metabolic Networks and Pathways
3.
Biochim Biophys Acta Rev Cancer ; 1877(5): 188785, 2022 09.
Article in English | MEDLINE | ID: mdl-36031088

ABSTRACT

Metabolic reprogramming is a unique but complex biochemical adaptation that allows solid tumors to tolerate various stresses that challenge cancer cells for survival. Under conditions of metabolic stress, mammalian cells employ adenosine monophosphate (AMP)-activated protein kinase (AMPK) to regulate energy homeostasis by controlling cellular metabolism. AMPK has been described as a cellular energy sensor that communicates with various metabolic pathways and networks to maintain energy balance. Earlier studies characterized AMPK as a tumor suppressor in the context of cancer. Later, a paradigm shift occurred in support of the oncogenic nature of AMPK, considering it a contextual oncogene. In support of this, various cellular and mouse models of tumorigenesis and clinicopathological studies demonstrated increased AMPK activity in various cancers. This review will describe AMPK's pro-tumorigenic activity in various malignancies and explain the rationale and context for using AMPK inhibitors in combination with anti-metabolite drugs to treat AMPK-driven cancers.


Subject(s)
AMP-Activated Protein Kinases , Neoplasms , AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/therapeutic use , Animals , Energy Metabolism/physiology , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Oncogenes/genetics , Phosphorylation
4.
FEBS J ; 289(6): 1575-1590, 2022 03.
Article in English | MEDLINE | ID: mdl-34668648

ABSTRACT

Hematopoietic PBX-interacting protein (HPIP, also known as PBXIP1) is an estrogen receptor (ER) interacting protein that regulates estrogen-mediated breast cancer cell proliferation and tumorigenesis. However, its functional significance in the context of mammary gland development is unexplored. Here, we report that HPIP is required for prolactin (PRL)-induced lactogenic differentiation in vitro. Molecular analysis of HPIP expression in mice revealed its induced expression at pregnancy and lactation stages of mammary gland. Moreover, PRL is a lactogenic hormone that controls pregnancy as well as lactation and induces Hpip/Pbxip1 expression in a signal transducer and activator of transcription 5a-dependent manner. Using mammary epithelial and lactogenic-competent cell lines, we further show that HPIP plays a regulatory role in PRL-mediated mammary epithelial cell differentiation, which is measured by acini formation, ß-casein synthesis, and lipid droplet formation. Further mechanistic studies using pharmacological inhibitors revealed that HPIP modulates PRL-induced ß-casein synthesis via phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) activation. This study also identified HPIP as a critical regulator of autocrine PRL signaling as treatment with the PRL receptor antagonist Δ1-9-G129R-hPRL restrained HPIP-mediated PRL synthesis, AKT activation, and ß-casein synthesis in cultured HC11 cells. Interestingly, we also uncovered that microRNA-148a (miR-148a) antagonizes HPIP-mediated mammary epithelial cell differentiation. Together, our study identified HPIP as a critical regulator of PRL signaling and revealed a novel molecular circuitry involving PRL, HPIP, PI3K/AKT, and miR-148a that controls mammary epithelial cell differentiation in vitro.


Subject(s)
MicroRNAs , Proto-Oncogene Proteins c-akt , Animals , Caseins/genetics , Caseins/metabolism , Cell Differentiation , Co-Repressor Proteins , Epithelial Cells/metabolism , Female , Mammary Glands, Animal , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Prolactin/genetics , Prolactin/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
5.
Cancer Lett ; 526: 12-28, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34767928

ABSTRACT

While phenotypic plasticity is a critical factor contributing to tumor heterogeneity, molecular mechanisms underlying this process are largely unknown. Here we report that breast cancer cells display phenotypic diversity in response to hypoxia or normoxia microenvironments by operating a reciprocal positive feedback regulation of HPIP and HIF-1α. We show that under hypoxia, HIF-1α induces HPIP expression that establishes cell survival, and also promotes cell migration/invasion, EMT and metastatic phenotypes in breast cancer cells. Mechanistic studies revealed that HPIP interacts with SRP14, a component of signal recognition particle, and stimulates MMP9 synthesis under hypoxic stress. Whereas, in normoxia, HPIP stabilizes HIF-1α, causing the Warburg effect to support cell growth. Concurrently, mathematical modelling corroborates this reciprocal feedback loop in enabling cell-state transitions in cancer cells. Clinical data indicate that elevated levels of HPIP and HIF-1α correlate with unfavorable prognosis and shorter survival rates in breast cancer subjects. Together, this data shows a reciprocal positive feedback loop between HPIP and HIF-1α that was unknown hitherto. It unveils how the tumor microenvironment influences phenotypic plasticity that has an impact on tumor growth and metastasis and, further signifies considering this pathway as a potential therapeutic target in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Phenotype
6.
Cancer Lett ; 518: 243-255, 2021 10 10.
Article in English | MEDLINE | ID: mdl-34302919

ABSTRACT

While cancer cells rewire metabolic pathways to sustain growth and survival under metabolic stress in solid tumors, the molecular mechanisms underlying these processes remain largely unknown. In this study, cancer cells switched from survival to death during the early to late phases of metabolic stress by employing a novel signaling switch from AMP activated protein kinase (AMPK)-Forkhead box O3 (FOXO3a)-hematopoietic PBX1-interacting protein (HPIP) to the ring finger protein 2 (RNF2)-HPIP-ubiquitin (Ub) pathway. Acute metabolic stress induced proto-oncogene HPIP expression in an AMPK-FOXO3a-dependent manner in breast cancer (BC) cells. HPIP depletion reduced cell survival and tumor formation in mouse xenografts, which was accompanied by diminished intracellular ATP levels and increased apoptosis in BC cells in response to metabolic (glucose) stress. Glutamine flux (13C-labeled) analysis further suggested that HPIP rewired glutamine metabolism by controlling the expression of the solute carrier family 1 member 5 (SLC1A5) and glutaminase (GLS) genes by acting as a coactivator of MYC to ensure cell survival upon glucose deprivation. However, in response to chronic glucose stress, HPIP was ubiquitinated by the E3-Ub ligase, RNF2, and was concomitantly degraded by the proteasome-mediated pathway, ensuring apoptosis. In support of these data, clinical analyses further indicated that elevated levels of HPIP correlated with AMPK activation in BC. Taken together, these data suggest that HPIP is a signal coordinator during metabolic stress and thus serves as a potential therapeutic target in BC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/metabolism , Glucose/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Adaptation, Physiological/physiology , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Line , Cell Line, Tumor , Female , Glutaminase/metabolism , Glutamine/metabolism , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Minor Histocompatibility Antigens , Stress, Physiological/physiology , Ubiquitin-Protein Ligases/metabolism
7.
Biomed Mater ; 15(5): 055025, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32464611

ABSTRACT

Zirconia-containing wollastonite (CaSiO3) ceramics with partial substitution of zirconia (1, 3 and 5 mol%) were prepared using eggshells and rice husk ash as source materials for calcium oxide and silica, respectively, through a sol-gel technique. The effect of incorporation of zirconia on in vitro bioactivity, mechanical properties, degradability and cytocompatibility of wollastonite was studied. Bioactivity was evaluated by in vitro assay using simulated body fluid while degradability was tested in Tris-HCl buffer solution for different time periods (1, 3, 7, 14 and 21 d) according to the ISO 10 993-14 standard. Human osteosarcoma (MG-63) cells were used to assess cytocompatibility with the MTT assay. X-ray diffractometry, Fourier transform infrared spectroscopy and scanning electron microscopy-energy dispersive spectroscopy were used to characterize the ceramics before and after in vitro studies. The results obtained showed that increasing the zirconia content in the wollastonite phase increases microhardness, compressive strength, bending strength and the elasticity modulus, while slightly decreasing the rate of formation of the hydroxyapatite layer. Moreover, the samples doped with zirconia had a lower degradation rate and it was noticed that cell viability is unaffected by the incorporation of zirconia.


Subject(s)
Biocompatible Materials/chemistry , Calcium Compounds/chemistry , Silicates/chemistry , Tissue Engineering/methods , Zirconium/chemistry , Buffers , Cell Line, Tumor , Cell Survival/drug effects , Ceramics , Durapatite/chemistry , Humans , In Vitro Techniques , Materials Testing , Microscopy, Electron, Scanning , Osteosarcoma/drug therapy , Spectroscopy, Fourier Transform Infrared , Tetrazolium Salts/pharmacology , Thermogravimetry , Thiazoles/pharmacology , X-Ray Diffraction
8.
Chem Biol Interact ; 308: 1-10, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31071337

ABSTRACT

Diarylheptanoids display an array of biological and pharmacological properties. We previously reported the synthesis of a diarylheptanoid Alpinoid c and a series of its derivatives, evaluated their cytotoxicity against various human cancer cells. We found some of these derivatives were significantly more potent than Alpinoid c in preventing the proliferation of various cancer cell lines. Among these, (S, E)-1-(3, 4 dimethoxyphenyl)-6-hydroxy-7-phenylhept-4-en-3-one (DPHP) showed most potent cytotoxicity against COLO205 cells, however, the mechanism by which DPHP prevents the growth of these colon cancer cells remains unknown. In the current study, we investigated the molecular mechanism of DPHP on colon cancer cells. DPHP inhibited the proliferation of COLO205 (IC50 7.01 ±â€¯0.62 µM) and A549 (IC50 20.03 ±â€¯3.11 µM) cells more specifically than normal human colon epithelial cell line NCM460 (IC50 55.6 ±â€¯4.02 µM). In COLO205 cells, DPHP induced cell shrinkage, membrane blebbing, chromatin condensation, phosphatidylserine externalization, and an accumulation of cells at sub-G1 phase. Further analysis these cells treated with DPHP revealed a decrease in mitochondrial membrane potential, an increase in Bax/Bcl2 ratio, the release of cytochrome c, activation of caspases -9, -3/7, and cleavage of the poly-ADP-ribose polymerase. DPHP treatment resulted in inhibition of hypoxia induced VEGF downstream signaling pathway in COLO205 cells is concurrent with inhibition of angiogenesis in CAM. Based on these data we suggest that DPHP significantly induced apoptosis possibly via intrinsic mitochondrial apoptosis pathway and inhibited angiogenesis. Our study suggests DPHP could be a therapeutic agent in treating colon cancer.


Subject(s)
Apoptosis/drug effects , Diarylheptanoids/pharmacology , Neovascularization, Physiologic/drug effects , A549 Cells , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Chick Embryo , Diarylheptanoids/chemistry , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
9.
J Biol Chem ; 294(26): 10236-10252, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31101654

ABSTRACT

Proper cell division relies on the coordinated regulation between a structural component, the mitotic spindle, and a regulatory component, anaphase-promoting complex/cyclosome (APC/C). Hematopoietic PBX-interacting protein (HPIP) is a microtubule-associated protein that plays a pivotal role in cell proliferation, cell migration, and tumor metastasis. Here, using HEK293T and HeLa cells, along with immunoprecipitation and immunoblotting, live-cell imaging, and protein-stability assays, we report that HPIP expression oscillates throughout the cell cycle and that its depletion delays cell division. We noted that by utilizing its D box and IR domain, HPIP plays a dual role both as a substrate and inhibitor, respectively, of the APC/C complex. We observed that HPIP enhances the G2/M transition of the cell cycle by transiently stabilizing cyclin B1 by preventing APC/C-Cdc20-mediated degradation, thereby ensuring timely mitotic entry. We also uncovered that HPIP associates with the mitotic spindle and that its depletion leads to the formation of multiple mitotic spindles and chromosomal abnormalities, results in defects in cytokinesis, and delays mitotic exit. Our findings uncover HPIP as both a substrate and an inhibitor of APC/C-Cdc20 that maintains the temporal stability of cyclin B1 during the G2/M transition and thereby controls mitosis and cell division.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdc20 Proteins/metabolism , Cell Cycle , Cyclin B1/chemistry , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Mitosis , Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Anaphase-Promoting Complex-Cyclosome/genetics , Cdc20 Proteins/antagonists & inhibitors , Cdc20 Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Spindle Apparatus , Substrate Specificity
11.
Drug Res (Stuttg) ; 69(7): 406-414, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30654398

ABSTRACT

Two series of diaziridinyl quinone isoxazole derivatives were prepared and evaluated for their cytotoxic activity against MCF7, HeLa, BT549, A549 and HEK293 cell lines and interaction with tubulin. Compounds (6A-M: ) showed promising activity against all the 5 human cancer cell lines. Compounds 6A: , 6E: and 6 M: were potent [IC50 ranging between 2.21 µg to 2.87 µg] on ER-positive MCF7 cell line similar to the commercially available drug molecule Doxorubicin. The results from docking models are in consistent with the experimental values which demonstrated the favourable binding modes of compounds 6A-M: to the interface of α- and ß-tubulin dimer.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Azirines/chemical synthesis , Azirines/pharmacology , Cell Line, Tumor , Chemistry Techniques, Synthetic , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Inhibitory Concentration 50 , Isoxazoles/chemical synthesis , Isoxazoles/pharmacology , Quinones/chemical synthesis , Quinones/pharmacology , Toxicity Tests , Tubulin/metabolism , Tubulin Modulators/chemical synthesis
12.
Biomed Mater ; 13(1): 015013, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29216013

ABSTRACT

Self-assembly is an important auto-organization process used in designing structural biomaterials which have the potential capability to heal tissues after traumatic injury. Although various materials having the ability to heal after injury are available, there is still a substantial need to develop new improved materials. To address this issue, we have developed hierarchical three-dimensional (3D) self-assembled zinc phosphate (Zn3(PO4)2) in the presence of cowpea mosaic virus (CPMV). Zn3(PO4)2 nanoparticles are self-assembled into nanosheets with a high degree of isotropy and then self-organized into a 3D structure that can enhance surface interactions with biological entities. The self-assembled structure is formed through the auto-organization of nanoparticles of size ∼50 nm under the influence of CPMV. The cellular response of self-assembled Zn3(PO4)2 and cell-particle adhesion behavior have been investigated through in vitro studies using modeled osteoblast-like MG63 cells. Self-assembled Zn3(PO4)2 resulted in proliferation of MG63 cells of up to 310% within 7 days of incubation. A 15% higher proliferation was obtained than with commercially available hydroxyapatite (HAp). Immunofluorescent analysis of MG63 cells after co-culturing with self-assembled Zn3(PO4)2 confirmed the healthy cytoskeletal organization and dense proliferation of MG63 cells. Further, Zn3(PO4)2 exhibited ∼28% cell-cycle progression in S phase, which is higher than obtained with commercially available HAp. Overall, these results demonstrate the multiple functions of hierarchical self-assembled Zn3(PO4)2 in the regeneration of bone tissue without defects and increasing the formation of cellular networks, and suggest its use in bone tissue engineering.


Subject(s)
Comovirus , Durapatite/chemistry , Metal Nanoparticles/chemistry , Phosphates/chemistry , Regeneration , Tissue Engineering/methods , Zinc Compounds/chemistry , Biocompatible Materials/chemistry , Bone and Bones , Cell Adhesion , Cell Culture Techniques , Cell Cycle , Cell Proliferation , Cell Survival , Coculture Techniques , Humans , Nanostructures , Osteoblasts/metabolism , Surface Properties
13.
Biomed Pharmacother ; 95: 956-964, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28915537

ABSTRACT

Remarkably, majority of the cancer deaths are due to metastasis, not because of primary tumors. Metastasis is one of the important hallmarks of cancer. During metastasis invasion of primary tumor cells from the site of origin to a new organ occurs. Metastasis associated proteins (MTAs) are a small family of transcriptional coregulators that are closely associated with tumor metastasis. These proteins are integral components of nuclear remodeling and deacetylation complex (NuRD). By virtue of being integral components of NuRD, these proteins regulate the gene expression by altering the epigenetic changes such as acetylation and methylation on the target gene chromatin. Among the MTA proteins, MTA1 expression is very closely correlated with the aggressiveness of several cancers that includes breast, liver, colon, pancreas, prostate, blood, esophageal, gastro-intestinal etc. Considering its close association with aggressiveness in human cancers, MTA1 may be considered as a potential therapeutic target for cancer treatment. The recent developments in its crystal structure further strengthened the idea of developing small molecule inhibitors for MTA1. In this review, we discuss the recent trends on the diverse functions of MTA1 and its role in various cancers, with the focus to consider MTA1 as a 'druggable' target in the control of human cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Repressor Proteins/genetics , Animals , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Models, Biological , Molecular Targeted Therapy , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Trans-Activators
14.
Mater Sci Eng C Mater Biol Appl ; 77: 772-779, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28532092

ABSTRACT

The present study aims to elucidate the applications of Titania (TiO2) doped calcium borosilicate glass as a biocompatible material in regenerative orthopedic applications. In this context, we have examined the bioactivity of various concentrations of TiO2 doped glasses with the help of simulated body fluid (SBF). Cytocompatibility, cell proliferation, and protein expression studies revealed the potential candidature of TiO2 doped glasses on osteoblast cell lines (MG-63). We hypothesized that TiO2 doped calcium borosilicate glasses are most cytocompatible material for bone implants. Glasses with composition 31B2O3-20SiO2-24.5Na2O-(24.5-x) CaO- x TiO2 (x=0,0.5,1,2) have been prepared by the conventional melt-quenching technique. After immersion of glasses in the SBF, formation of hydroxyapatite layer on the surface was confirmed by X-ray Diffractometer (XRD), Fourier Transform Infrared Spectroscopy (FT-IR) and Scanning Electron Microscopy-Energy Dispersive Spectroscopy (SEM-EDS) analysis. Significant change in the pH of the body fluid was observed with the addition of titania. Degradation test was performed as per the ISO 10993. The results showed that partial substitution of TiO2 with CaO negatively influenced bioactivity; it decreased with increase in concentration of TiO2. Vickers hardness tester was used to measure the microhardness values of the prepared glasses. With the increasing of TiO2 content, the microhardness of the glass samples was increased from 545Hv to 576Hv. Cytocompatibility has been evaluated with MG-63 cells by using MTT assay. Further, we observed that there was no change in expressions of cyclin levels even after the incorporation of titania. The antibacterial properties were examined against E. coli and S. aureus. Strong antibacterial efficacy was observed for 2% TiO2 in the system. Hence it can be concluded that titania-doped borosilicate glasses may be used as potential materials in bone tissue engineering.


Subject(s)
Glass , Biocompatible Materials , Calcium , Escherichia coli , Humans , Materials Testing , Spectroscopy, Fourier Transform Infrared , Staphylococcus aureus , Titanium , X-Ray Diffraction
15.
Cell Oncol (Dordr) ; 40(2): 133-144, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28039608

ABSTRACT

PURPOSE: Hematopoietic PBX interacting protein (HPIP), a scaffold protein, is known to regulate the proliferation, migration and invasion in different cancer cell types. The aim of this study was to assess the role of HPIP in ovarian cancer cell migration, invasion and epithelial-mesenchymal transition (EMT), and to unravel the mechanism by which it regulates these processes. METHODS: HPIP expression was assessed by immunohistochemistry of tissue microarrays containing primary ovarian tumor samples of different grades. OAW42, an ovarian carcinoma-derived cell line exhibiting a high HPIP expression, was used to study the role of HPIP in cell migration, invasion and EMT. HPIP knockdown in these cells was achieved using a small hairpin RNA (shRNA) approach. Cell migration and invasion were assessed using scratch wound and transwell invasion assays, respectively. The extent of EMT was assessed by determining the expression levels of Snail, Vimentin and E-cadherin using Western blotting. The effect of HPIP expression on AKT and MAPK activation was also investigated by Western blotting. Cell viabilities in response to cisplatin treatment were assessed using a MTT assay, whereas apoptosis was assessed by determining caspase-3 and PARP cleavage in ovarian carcinoma-derived SKOV3 cells. RESULTS: We found that HPIP is highly expressed in high-grade primary ovarian tumors. In addition, we found that HPIP promotes the migration, invasion and EMT in OAW42 cells and induces EMT in these cells via activation of the PI3K/AKT pathway. The latter was found to lead to stabilization of the Snail protein and to repression of E-cadherin expression through inactivation of GSK-3ß. We also found that HPIP expression confers cisplatin resistance to SKOV3 cells after prolonged exposure and that its subsequent knockdown decreases the viability of these cells and increases caspase-3 activation and PARP proteolysis in these cells following cisplatin treatment. CONCLUSIONS: From these results we conclude that HPIP expression is associated with high-grade ovarian tumors and may promote their migration, invasion and EMT, a process that is associated with metastasis. In addition, we conclude that HPIP may serve as a potential therapeutic target for cisplatin resistant ovarian tumors.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Neoplasm Grading , Neoplasm Invasiveness , Ovarian Neoplasms/enzymology , Protein Stability/drug effects , Snail Family Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...