Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Protein Sci ; 31(11): e4468, 2022 11.
Article in English | MEDLINE | ID: mdl-36214056

ABSTRACT

The vaccinia virus expression system is known for the efficient production of recombinant proteins with "appropriate" posttranslational modification using desired mammalian cell lines. However, being a replication competent virus, vaccinia virus poses a health threat to immunocompromised individuals and requires biosafety level 2 (BSL2) laboratory precautions, thereby restricting its use by the scientific community. Development of the host range restricted modified vaccinia Ankara (MVA) system has allowed researchers to work with a safer virus even at BSL1. Here, we report on the use of an improved second generation MVA viral system incorporating two selective markers and fluorescent proteins for easier recombinant virus identification. Notably, we demonstrate that this novel system is capable of producing secreted recombinant proteins, a finding not previously reported. Through purification and characterization of wild type and mutant platelet-derived growth factor D (PDGF D) dimer species, we demonstrate this system is capable of producing the latent full-length PDGF D dimer, partially processed intermediate dimer (hemidimer), as well as fully processed growth factor domain dimer that show chemical integrity and biological activity. Importantly, this system is amenable to scaling up for the mass production of recombinant PDGF D (rPDGF D) dimer species.


Subject(s)
Vaccinia virus , Vaccinia , Humans , Animals , Vaccinia virus/genetics , Virus Replication , Platelet-Derived Growth Factor , Recombinant Proteins/genetics , Mammals
2.
J Cell Physiol ; 237(11): 4180-4196, 2022 11.
Article in English | MEDLINE | ID: mdl-35994698

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces death receptor-mediated extrinsic apoptosis, specifically in cancer cells, and Bid (BH3-interacting domain death agonist) plays an important role in TRAIL-induced apoptosis. Ferroptosis is a newly defined form of regulated cell death known to be distinct from other forms of cell death. However, our previous studies have shown that ferroptosis shares common pathways with other types of programmed cell death such as apoptosis. In this study, we investigated the role of Bid in the crosstalk between the ferroptotic agent-induced endoplasmic reticulum (ER) stress response and TRAIL-induced apoptosis. When human colorectal carcinoma HCT116 cells were treated with the ferroptosis-inducing agents artesunate and erastin in combination with TRAIL, TRAIL-induced activation of caspase-8 was enhanced, and subsequently, the truncation of Bid was increased. Similar results were observed when ovarian adenocarcinoma OVCAR-3 cells were treated with the ferroptotic agents in combination with TRAIL. Results from studies with Bid mutants reveal that the truncation of Bid and the presence of intact BH3 domains are critical for synergistic apoptosis. Nonfunctional Bid mutants were not able to activate the mitochondria-dependent apoptosis pathway, which is required for the conversion of p19 to p17, the active form of caspase-3. These results indicate that Bid plays a critical role in the crosstalk between the ferroptotic agent-induced ER stress response and TRAIL-induced apoptosis.


Subject(s)
Apoptosis , Ovarian Neoplasms , Humans , Female , Cell Line, Tumor , Ovarian Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 8/metabolism , Endoplasmic Reticulum Stress , Tumor Necrosis Factor-alpha/metabolism
3.
J Biol Chem ; 298(6): 101981, 2022 06.
Article in English | MEDLINE | ID: mdl-35472332

ABSTRACT

Mesenchymal stem cells (MSCs) are adult stem cell populations and exhibit great potential in regenerative medicine and oncology. Platelet-derived growth factors (PDGFs) are well known to regulate MSC biology through their chemotactic and mitogenic properties. However, their direct roles in the regulation of MSC lineage commitment are unclear. Here, we show that PDGF D promotes the differentiation of human bone marrow mesenchymal stem cells (hBMSCs) into osteoblasts and inhibits hBMSC differentiation into adipocytes. We demonstrate that PDGF D-induced ß-actin expression and polymerization are essential for mediating this differential regulation of osteoblastogenesis and adipogenesis. Interestingly, we found that PDGF D induces massive upward molecular weight shifts of its cognate receptor, PDGF receptor beta (ß-PDGFR) in hBMSCs, which was not observed in fibroblasts. Proteomic analysis indicated that the E3 ubiquitin ligase HECT, UBA, and WWE domain-containing protein 1 (HUWE1) associates with the PDGF D-activated ß-PDGFR signaling complex in hBMSCs, resulting in ß-PDGFR polyubiquitination. In contrast to the well-known role of ubiquitin in protein degradation, we provide evidence that HUWE1-mediated ß-PDGFR polyubiquitination delays ß-PDGFR internalization and degradation, thereby prolonging AKT signaling. Finally, we demonstrate that HUWE1-regulated ß-PDGFR signaling is essential for osteoblastic differentiation of hBMSCs, while being dispensable for PDGF D-induced hBMSC migration and proliferation as well as PDGF D-mediated inhibition of hBMSC differentiation into adipocytes. Taken together, our findings provide novel insights into the molecular mechanism by which PDGF D regulates the commitment of hBMSCs into the osteoblastic lineage.


Subject(s)
Lymphokines/metabolism , Mesenchymal Stem Cells , Platelet-Derived Growth Factor/metabolism , Ubiquitin-Protein Ligases , Cell Differentiation , Cell Proliferation , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Proteomics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
4.
Cells ; 10(10)2021 10 12.
Article in English | MEDLINE | ID: mdl-34685701

ABSTRACT

A hallmark of malignant solid tumor is extracellular acidification coupled with metabolic switch to aerobic glycolysis. Using the human MCF10A progression model of breast cancer, we show that glycolytic switch and extracellular acidosis in aggressive cancer cells correlate with increased expression of tissue inhibitor of metalloproteinase-1 (TIMP-1), known to induce intracellular signal transduction through the interaction with its cell surface receptor CD63, independent of its metalloproteinase inhibitory function. We found that, in aggressive breast carcinoma, the TIMP-1-CD63 signaling axis induced a metabolic switch by upregulating the rate of aerobic glycolysis, lowering mitochondrial respiration, preventing intracellular acidification, and inducing extracellular acidosis. Carbonic anhydrase IX (CAIX), a regulator of cellular pH through the hydration of metabolically released pericellular CO2, was identified as a downstream mediator of the TIMP-1-CD63 signaling axis responsible for extracellular acidosis. Consistently with our previous study, the TIMP-1-CD63 signaling promoted survival of breast cancer cells. Interestingly, breast carcinoma cell survival was drastically reduced upon shRNA-mediated knockdown of CAIX expression, demonstrating the significance of CAIX-regulated pH in the TIMP-1-CD63-mediated cancer cell survival. Taken together, the present study demonstrates the functional significance of TIMP-1-CD63-CAXI signaling axis in the regulation of tumor metabolism, extracellular acidosis, and survival of breast carcinoma. We propose that this axis may serve as a novel therapeutic target.


Subject(s)
Breast Neoplasms/metabolism , Tetraspanin 30/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Acids/metabolism , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/metabolism , Cell Line, Tumor , Cell Survival , Disease Progression , Extracellular Space/metabolism , Female , Humans , Models, Biological , Neoplasm Invasiveness
5.
Cell Death Dis ; 12(11): 997, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34697296

ABSTRACT

The autophagy-lysosome pathway and apoptosis constitute vital determinants of cell fate and engage in a complex interplay in both physiological and pathological conditions. Central to this interplay is the archetypal autophagic cargo adaptor p62/SQSTM1/Sequestosome-1 which mediates both cell survival and endoplasmic reticulum stress-induced apoptosis via aggregation of ubiquitinated caspase-8. Here, we investigated the role of p62-mediated apoptosis in head and neck squamous cell carcinoma (HNSCC), which can be divided into two groups based on human papillomavirus (HPV) infection status. We show that increased autophagic flux and defective apoptosis are associated with radioresistance in HPV(-) HNSCC, whereas HPV(+) HNSCC fail to induce autophagic flux and readily undergo apoptotic cell death upon radiation treatments. The degree of radioresistance and tumor progression of HPV(-) HNSCC respectively correlated with autophagic activity and cytosolic levels of p62. Pharmacological activation of the p62-ZZ domain using small molecule ligands sensitized radioresistant HPV(-) HNSCC cells to ionizing radiation by facilitating p62 self-polymerization and sequestration of cargoes leading to apoptosis. The self-polymerizing activity of p62 was identified as the essential mechanism by which ubiquitinated caspase-8 is sequestered into aggresome-like structures, without which irradiation fails to induce apoptosis in HNSCC. Our results suggest that harnessing p62-dependent sequestration of ubiquitinated caspase-8 provides a novel therapeutic avenue in patients with radioresistant tumors.


Subject(s)
Apoptosis/immunology , Radiation, Ionizing , Sequestosome-1 Protein/metabolism , Animals , Caspase 8 , Humans , Mice , Radiation Injuries , Signal Transduction
6.
Cancers (Basel) ; 13(6)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801879

ABSTRACT

Despite recent advances in therapeutic modalities such as radiochemotherapy, the long-term prognosis for patients with advanced head and neck squamous cell carcinoma (HNSCC), especially nonviral HNSCC, remains very poor, while survival of patients with human papillomavirus (HPV)-associated HNSCC is greatly improved after radiotherapy. The goal of this study is to develop a mechanism-based treatment protocol for high-risk patients with HPV-negative HNSCC. To achieve our goal, we have investigated molecular mechanisms underlying differential radiation sensitivity between HPV-positive and -negative HNSCC cells. Here, we found that autophagy is associated with radioresistance in HPV-negative HNSCC, whereas apoptosis is associated with radiation sensitive HPV-positive HNSCC. Interestingly, we found that photodynamic therapy (PDT) directed at the endoplasmic reticulum (ER)/mitochondria initially induces paraptosis followed by apoptosis. This led to a substantial increase in radiation responsiveness in HPV-negative HNSCC, while the same PDT treatment had a minimal effect on HPV-positive cells. Here, we provide evidence that the autophagic adaptor p62 mediates signal relay for the induction of apoptosis, promoting ionizing radiation (XRT)-induced cell death in HPV-negative HNSCC. This work proposes that ER/mitochondria-targeted PDT can serve as a radiosensitizer in intrinsically radioresistant HNSCC that exhibits an increased autophagic flux.

7.
Sci Rep ; 10(1): 2099, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034211

ABSTRACT

Tissue inhibitor of metalloproteinases-1 (TIMP-1) is a pleiotropic protein, promoting both tumor-suppressive and tumor-promoting activities. While TIMP-1 is primarily known as an endogenous inhibitor of matrix metalloproteinases (MMPs) and thus associated with tumor cell invasion, clinical studies demonstrated increased expression of TIMP-1 and its association with poor prognosis in cancer. Non-MMP-inhibitory and oncogenic functions of TIMP-1 are mediated by induction of intracellular signaling via its cell surface receptor CD63, a tetraspanin. The present study investigates the structure-function relationship of TIMP-1 for its interaction with CD63, which may eventually help design a novel approach for targeting TIMP-1's pro-oncogenic activity without interfering its tumor suppressive MMP-inhibitory function. Importantly, our analysis includes TIMP-1/CD63 interactions at the cell surface of live cells. Here, we demonstrate that the 9 C-terminal amino acid residues of TIMP-1 and the large extracellular loop of CD63 are required for their interaction. Considering that the N-terminal half of TIMP-1 is sufficient for TIMP-1's MMP-inhibitory activity, we propose that those C-terminal amino acid residues are a potentially targetable motif of TIMP-1 oncogenic activity. As a proof of concept, we present the potential for the development of neutralizing antibodies against the C-terminal motif of TIMP-1 for disruption of TIMP-1 interaction with CD63 and the subsequent signal transduction.


Subject(s)
Neoplasms/metabolism , Tetraspanin 30/metabolism , Tissue Inhibitor of Metalloproteinase-1/chemistry , HEK293 Cells , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Structure-Activity Relationship , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/physiology , Two-Hybrid System Techniques
8.
Growth Factors ; 37(3-4): 131-145, 2019 08.
Article in English | MEDLINE | ID: mdl-31542979

ABSTRACT

Despite strong evidence for the involvement of PDGF signaling in breast cancer, little is known about the PDGF ligand responsible for PDGFR activation during breast cancer progression. Here, we found PDGF-C to be highly expressed in breast carcinoma cell lines. Immunohistochemical analysis of invasive breast cancer revealed an association between increased PDGF-C expression and lymph node metastases, Ki-67 proliferation index, and poor disease-free survival. We also identified a PDGF-C splice variant encoding truncated PDGF-C (t-PDGF-C) isoform lacking the signal peptide and the N-terminal CUB domain. While t-PDGF C homodimer is retained intracellularly, it can be secreted as a heterodimer with full-length PDGF-C (FL-PDGF-C). PDGF-C downregulation reduced anchorage-independent growth and matrigel invasion of MDA-MB-231 cells. Conversely, ectopic expression of t-PDGF-C enhanced phenotypic transformation and invasion in BT-549 cells expressing endogenous FL-PDGF-C. The present study provides new insights into the functional significance of PDGF-C and its splice variant in human breast cancer.


Subject(s)
Breast Neoplasms/pathology , Lymphatic Metastasis/genetics , Lymphokines/genetics , Lymphokines/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Lymphatic Metastasis/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction
9.
Oncotarget ; 8(31): 51530-51541, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881665

ABSTRACT

HPV-positive oropharyngeal cancer patients experience significantly lower locoregional recurrence and higher overall survival in comparison with HPV-negative patients, especially among those who received radiation therapy. The goal of the present study is to investigate the molecular mechanisms underlying the differential radiation sensitivity between HPV-negative and HPV-positive head and neck squamous cell carcinoma (HNSCC). Here, we show that HPV-negative HNSCC cells exhibit increased glucose metabolism as evidenced by increased production of lactate, while HPV-positive HNSCC cells effectively utilize mitochondrial respiration as evidenced by increased oxygen consumption. HPV-negative cells express HIF1α and its downstream mediators of glucose metabolism such as hexokinase II (HKII) and carbonic anhydrase IX (CAIX) at higher levels, while the expression level of cytochrome c oxidase (COX) was noticeably higher in HPV-positive HNSCC. In addition, the expression levels of pyruvate dehydrogenase kinases (PDKs), which inhibit pyruvate dehydrogenase activity, thereby preventing entry of pyruvate into the mitochondrial tricarboxylic acid (TCA) cycle, were much higher in HPV-negative HNSCC compared to those in HPV-positive cells. Importantly, a PDK inhibitor, dichloroacetate, effectively sensitized HPV-negative cells to irradiation. Lastly, we found positive interactions between tonsil location and HPV positivity for COX intensity and COX/HKII index ratio as determined by immunohistochemical analysis. Overall survival of patients with HNSCC at the tonsil was significantly improved with an increased COX expression. Taken together, the present study provides molecular insights into the mechanistic basis for the differential responses to radiotherapy between HPV-driven vs. spontaneous or chemically induced oropharyngeal cancer.

10.
J Transl Med ; 14: 72, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26975354

ABSTRACT

BACKGROUND: Characterization of genes linked to bone metastasis is critical for identification of novel prognostic or predictive biomarkers and potential therapeutic targets in metastatic castrate-resistant prostate cancer (mCRPC). Although bone marrow core biopsies (BMBx) can be obtained for gene profiling, the procedure itself is invasive and uncommon practice in mCRPC patients. Conversely, circulating tumor cells (CTCs), which are likely to stem from bone metastases, can be isolated from blood. The goals of this exploratory study were to establish a sensitive methodology to analyze gene expression in BMBx and CTCs, and to determine whether the presence or absence of detectable gene expression is concordant in matching samples from mCRPC patients. METHODS: The CellSearch(®) platform was used to enrich and enumerate CTCs. Low numbers of PC3 prostate cancer (PCa) cells were spiked into normal blood to assess cell recovery rate. RNA extracted from recovered PC3 cells was amplified using an Eberwine-based procedure to obtain antisense mRNA (aRNA), and assess the linearity of the RNA amplification method. In this pilot study, RNAs extracted from CTCs and PCa cells microdissected from formalin-fixed paraffin-embedded BMBx, were amplified to obtain aRNA and assess the expression of eight genes functionally relevant to PCa bone metastasis using RT-PCR. RESULTS: RNAs were successfully extracted from as few as 1-5 PCa cells in blood samples. The relative expression levels of reference genes were maintained after RNA amplification. The integrity of the amplified RNA was also demonstrated by RT-PCR analysis using primer sets that target the 5'-end, middle, and 3'-end of reference mRNA. We found that in 21 out of 28 comparisons, the presence or absence of detectable gene expression in CTCs and PCa cells microdissected from single bone lesions of the same patients was concordant. CONCLUSIONS: This exploratory analysis suggests that aRNA amplification through in vitro transcription may be useful as a method to detect gene expression in small numbers of CTCs and tumor cells microdissected from bone metastatic lesions. In some cases, gene expression in CTCs and BMBxs was not concordant, raising questions about using CTC gene expression to make clinical decisions.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/secondary , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Aged , Aged, 80 and over , Biopsy , Bone Marrow/pathology , Cell Line, Tumor , Humans , Male , Middle Aged , Prostatic Neoplasms, Castration-Resistant/genetics , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reproducibility of Results
11.
Prostate ; 76(6): 534-42, 2016 May.
Article in English | MEDLINE | ID: mdl-26732854

ABSTRACT

PURPOSE: To determine the functional relationship between androgen receptor (AR) and PDGF D as it relates to the radiation response of PTEN-null prostate cancer (PCa) cells and the effect of enzalutamide on these interactions. METHODS AND MATERIALS: Using murine PTEN-null prostate epithelial cell line and human prostate carcinoma LNCaP (PTEN-mutant) models, nuclear and cytosolic AR levels were determined by immunoblot analysis and the transcriptional activity of nuclear AR was assessed by RT-PCR analysis of its target genes with or without irradiation. Cell survival was evaluated by clonogenic assay or sulforhodamine B (SRB) assay upon irradiation in the absence or presence of the AR antagonist enzalutamide. RESULTS: PTEN loss resulted in upregulation of AR expression in a PDGF-D dependent manner and irradiation selectively increased the nuclear AR protein level and its activity in a murine cell model. When the functional significance of AR in cell survival was tested, treatment with enzalutamide resulted in radiosensitization of human LNCaP cells. Similarly to the murine model, PDGF-D overexpression increased the nuclear AR level and its transcriptional activity in LNCaP cells. PDGF-D over-expression was associated with radioresistance and enzalutamide treatment effectively reversed PDGF-D-mediated radioresistance in LNCaP cells. CONCLUSIONS: We have demonstrated that AR, a target of the PTEN and PDGF D-downstream signaling program, contributes to radiation resistance in human PCa cells. In addition, this study suggests that anti-androgens such as enzalutamide may serve as radiation sensitizers for the treatment of PCa patients, particularly so in patients with loss of PTEN or overexpression of PDGF-D.


Subject(s)
Lymphokines/metabolism , Phenylthiohydantoin/analogs & derivatives , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms , Receptors, Androgen/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Humans , Male , Mice , Nitriles , PTEN Phosphohydrolase/metabolism , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tumor Suppressor Proteins/metabolism
12.
Am J Physiol Cell Physiol ; 310(4): C293-304, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26157007

ABSTRACT

Activation of ß-platelet-derived growth factor receptor (ß-PDGFR) is associated with prostate cancer (PCa) progression and recurrence after prostatectomy. Analysis of the ß-PDGFR ligands in PCa revealed association between PDGF-D expression and Gleason score as well as tumor stage. During the course of studying the functional consequences of PDGF ligand-specific ß-PDGFR signaling in PCa, we discovered a novel function of PDGF-D for activation/shedding of the serine protease matriptase leading to cell invasion, migration, and tumorigenesis. The present study showed that PDGF-D, not PDGF-B, induces extracellular acidification, which correlates with increased matriptase activation. A cDNA microarray analysis revealed that PDGF-D/ß-PDGFR signaling upregulates expression of the acidosis regulator carbonic anhydrase IX (CAIX), a classic target of the transcriptional factor hypoxia-inducible factor-1α (HIF-1α). Cellular fractionation displayed a strong HIF-1α nuclear localization in PDGF-D-expressing cells. Treatment of vector control or PDGF-B-expressing cells with the HIF-1α activator CoCl2 led to increased CAIX expression accompanied by extracellular acidosis and matriptase activation. Furthermore, the analysis of the CAFTD cell lines, variants of the BPH-1 transformation model, showed that increased PDGF-D expression is associated with enhanced HIF-1α activity, CAIX induction, cellular acidosis, and matriptase shedding. Importantly, shRNA-mediated knockdown of CAIX expression effectively reversed extracellular acidosis and matriptase activation in PDGF-D-transfected BPH-1 cells and in CAFTD variants that express endogenous PDGF-D at a high level. Taken together, these novel findings reveal a new paradigm in matriptase activation involving PDGF-D-specific signal transduction leading to extracellular acidosis.


Subject(s)
Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms/enzymology , Serine Endopeptidases/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Autocrine Communication , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Cell Line, Tumor , Enzyme Activation , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphokines/genetics , Male , Platelet-Derived Growth Factor/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Serine Endopeptidases/genetics , Signal Transduction , Time Factors , Transfection
13.
Mol Cancer Res ; 12(9): 1324-33, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24895412

ABSTRACT

UNLABELLED: Tissue inhibitor of metalloproteinase-1 (TIMP-1) regulates intracellular signaling networks for inhibition of apoptosis. Tetraspanin (CD63), a cell surface binding partner for TIMP-1, was previously shown to regulate integrin-mediated survival pathways in the human breast epithelial cell line MCF10A. In the current study, we show that TIMP-1 expression induces phenotypic changes in cell morphology, cell adhesion, cytoskeletal remodeling, and motility, indicative of an epithelial-mesenchymal transition (EMT). This is evidenced by loss of the epithelial cell adhesion molecule E-cadherin with an increase in the mesenchymal markers vimentin, N-cadherin, and fibronectin. Signaling through TIMP-1, but not TIMP-2, induces the expression of TWIST1, an important EMT transcription factor known to suppress E-cadherin transcription, in a CD63-dependent manner. RNAi-mediated knockdown of TWIST1 rescued E-cadherin expression in TIMP-1-overexpressing cells, demonstrating a functional significance of TWIST1 in TIMP-1-mediated EMT. Furthermore, analysis of TIMP-1 structural mutants reveals that TIMP-1 interactions with CD63 that activate cell survival signaling and EMT do not require the matrix metalloproteinase (MMP)-inhibitory domain of TIMP-1. Taken together, these data demonstrate that TIMP-1 binding to CD63 activates intracellular signal transduction pathways, resulting in EMT-like changes in breast epithelial cells, independent of its MMP-inhibitory function. IMPLICATIONS: TIMP-1's function as an endogenous inhibitor of MMP or as a "cytokine-like" signaling molecule may be a critical determinant for tumor cell behavior.


Subject(s)
Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Nuclear Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Twist-Related Protein 1/metabolism , Apoptosis/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation/genetics , Cell Survival/genetics , Epithelial Cells , Female , Gene Expression Regulation, Neoplastic , Humans , Nuclear Proteins/genetics , Signal Transduction , Tetraspanin 30 , Tissue Inhibitor of Metalloproteinase-1/metabolism , Twist-Related Protein 1/genetics
14.
Int J Radiat Oncol Biol Phys ; 88(1): 151-8, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24331662

ABSTRACT

PURPOSE: Loss or mutation of the phosphate and tensin homologue (PTEN) is a common genetic abnormality in prostate cancer (PCa) and induces platelet-derived growth factor D (PDGF D) signaling. We examined the role of the PTEN/PDGF axis on radioresponse using a murine PTEN null prostate epithelial cell model. METHODS AND MATERIALS: PTEN wild-type (PTEN+/+) and PTEN knockout (PTEN-/-) murine prostate epithelial cell lines were used to examine the relationship between the PTEN status and radiosensitivity and also to modulate the PDGF D expression levels. PTEN-/- cells were transduced with a small hairpin RNA (shRNA) lentiviral vector containing either scrambled nucleotides (SCRM) or sequences targeted to PDGF D (shPDGF D). Tumorigenesis and morphogenesis of these cell lines were evaluated in vivo via subcutaneous injection of male nude mice and in vitro using Matrigel 3-dimensional (3D) culture. Effects of irradiation on clonogenic survival, cell migration, and invasion were measured with respect to the PTEN status and the PDGF D expression level. In addition, apoptosis and cell cycle redistribution were examined as potential mechanisms for differences seen. RESULTS: PTEN-/- cells were highly tumorigenic in animals and effectively formed foci in 3D culture. Importantly, loss of PDGF D in these cell lines drastically diminished these phenotypes. Furthermore, PTEN-/- cells demonstrated increased clonogenic survival in vitro compared to PTEN+/+, and attenuation of PDGF D significantly reversed this radioresistant phenotype. PTEN-/- cells displayed greater migratory and invasive potential at baseline as well as after irradiation. Both the basal and radiation-induced migratory and invasive phenotypes in PTEN-/- cells required PDGF D expression. Interestingly, these differences were independent of apoptosis and cell cycle redistribution, as they showed no significant difference. CONCLUSIONS: We propose that PDGF D represents a potentially promising target for PCa treatment resistance in the absence of PTEN function, and warrants further laboratory evaluation and clinical study.


Subject(s)
Adenocarcinoma/radiotherapy , Cell Transformation, Neoplastic , Lymphokines/physiology , Neoplasm Proteins/physiology , PTEN Phosphohydrolase/physiology , Platelet-Derived Growth Factor/physiology , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cell Line, Tumor , Cell Movement/radiation effects , Cell Survival/radiation effects , Cell Transformation, Neoplastic/pathology , Male , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , Neoplasm Proteins/deficiency , PTEN Phosphohydrolase/deficiency , Phenotype , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, Platelet-Derived Growth Factor beta , Signal Transduction/physiology
15.
Mol Cancer Res ; 10(8): 1087-97, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22689130

ABSTRACT

The platelet-derived growth factors (PDGF A, B, C, and D) and their receptors (α-PDGFR and ß-PDGFR) play an indispensible role in physiologic and pathologic conditions, including tumorigenesis. The transformative ß-PDGFR is overexpressed and activated during prostate cancer progression, but the identification and functional significance of its complementary ligand have not been elucidated. This study examined potential oncogenic functions of ß-PDGFR ligands PDGF B and PDGF D, using nonmalignant prostate epithelial cells engineered to overexpress these ligands. In our models, PDGF D induced cell migration and invasion more effectively than PDGF B in vitro. Importantly, PDGF D supported prostate epithelial cell tumorigenesis in vivo and showed increased tumor angiogenesis compared with PDGF B. Autocrine signaling analysis of the mitogen-activated protein kinase and phosphoinositide 3-kinase pathways found PDGF D-specific activation of the c-jun-NH2-kinase (JNK) signaling cascade. Using short hairpin RNA and pharmacologic inhibitors, we showed that PDGFD-mediated phenotypic transformation is ß-PDGFR and JNK dependent. Importantly, we made a novel finding of PDGF D-specific increase in the shedding and activation of the serine protease matriptase in prostate epithelial cells. Our study, for the first time to our knowledge, showed ligand-specific ß-PDGFR signaling as well as PDGF D-specific regulation of matriptase activity and its spatial distribution through shedding. Taken together with our previous finding that matriptase is a proteolytic activator of PDGF D, this study provides a molecular insight into signal amplification of the proteolytic network and PDGF signaling loop during cancer progression.


Subject(s)
Lymphokines , Platelet-Derived Growth Factor , Prostatic Neoplasms , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor beta , Animals , Becaplermin , Cell Line, Tumor , Cell Movement , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Lymphokines/genetics , Lymphokines/metabolism , Male , Mice , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Invasiveness , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction
16.
Prostate ; 72(12): 1328-38, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22213159

ABSTRACT

BACKGROUND: The major cause of death in prostate cancer (PCa) cases is due to distant metastatic lesions, with the bone being the most prevalent site for secondary colonization. Utilization of small molecule inhibitors to treat bone metastatic PCa have had limited success either as monotherapies or in combination with other chemotherapeutics due to intolerable toxicities. In the current study, we developed a clinically relevant in vivo intraosseous tumor model overexpressing the platelet-derived growth factor D (PDGF D) to test the efficacy of a newly characterized vascular endothelial growth factor receptor (VEGFR)/PDGFR inhibitor, cediranib (also called AZD2171). METHODS: An intratibial-injection model was established utilizing DU145 cells with or without increased PDGF D expression. Tumor-bearing mice were treated by daily gavage administration of cediranib and/or weekly i.p. injection of docetaxel for 7 weeks. Tibiae were monitored by in vivo/ex vivo X-rays and histomorphometry analysis was performed to estimate tumor volume and tumor-associated trabecular bone growth. RESULTS: Cediranib reduced intraosseous growth of prostate tumors as well as tumor-associated bone responses. When compared to the standard chemotherapeutic agent docetaxel, cediranib exhibited a stronger inhibition of tumor-associated bone response. The efficacy of cediranib was further enhanced when the drug was co-administered with docetaxel. Importantly, the therapeutic benefits of cediranib and docetaxel are more prominent in intraosseous prostate tumors overexpressing PDGF D. CONCLUSION: These novel findings support the utilization of cediranib, either alone or in combination with docetaxel, to treat bone metastatic PCa exhibiting PDGF D expression.


Subject(s)
Bone Neoplasms/drug therapy , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Growth Inhibitors/therapeutic use , Lymphokines/biosynthesis , Platelet-Derived Growth Factor/biosynthesis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Quinazolines/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Humans , Lymphokines/antagonists & inhibitors , Male , Mice , Mice, SCID , Platelet-Derived Growth Factor/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Random Allocation , Xenograft Model Antitumor Assays/methods
17.
Biochem J ; 441(3): 909-18, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22035541

ABSTRACT

The PDGF (platelet-derived growth factor) family members are potent mitogens for cells of mesenchymal origin and serve as important regulators of cell migration, survival, apoptosis and transformation. Tumour-derived PDGF ligands are thought to function in both autocrine and paracrine manners, activating receptors on tumour and surrounding stromal cells. PDGF-C and -D are secreted as latent dimers, unlike PDGF-A and -B. Cleavage of the CUB domain from the PDGF-C and -D dimers is required for their biological activity. At present, little is known about the proteolytic processing of PDGF-C, the rate-limiting step in the regulation of PDGF-C activity. In the present study we show that the breast carcinoma cell line MCF7, engineered to overexpress PDGF-C, produces proteases capable of cleaving PDGF-C to its active form. Increased PDGF-C expression enhances cell proliferation, anchorage-independent cell growth and tumour cell motility by autocrine signalling. In addition, MCF7-produced PDGF-C induces fibroblast cell migration in a paracrine manner. Interestingly, PDGF-C enhances tumour cell invasion in the presence of fibroblasts, suggesting a role for tumour-derived PDGF-C in tumour-stromal interactions. In the present study, we identify tPA (tissue plasminogen activator) and matriptase as major proteases for processing of PDGF-C in MCF7 cells. In in vitro studies, we also show that uPA (urokinase-type plasminogen activator) is able to process PDGF-C. Furthermore, by site-directed mutagenesis, we identify the cleavage site for these proteases in PDGF-C. Lastly, we provide evidence suggesting a two-step proteolytic processing of PDGF-C involving creation of a hemidimer, followed by GFD-D (growth factor domain dimer) generation.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Serine Proteases/physiology , Animals , Autocrine Communication/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Disease Progression , Female , Humans , Lymphokines/antagonists & inhibitors , Lymphokines/genetics , Lymphokines/physiology , Mice , NIH 3T3 Cells , Paracrine Communication/genetics , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/physiology , Protein Multimerization/genetics , Protein Multimerization/physiology , Protein Processing, Post-Translational , Serine Proteases/genetics , Serine Proteases/metabolism , Transfection
18.
Curr Pharm Des ; 15(20): 2311-8, 2009.
Article in English | MEDLINE | ID: mdl-19601833

ABSTRACT

ADAM15 is a widely expressed multi-domain protease that has been implicated in the pathogenesis of many human diseases. Given the diversity of the ADAM15 functional domains, this protease is thought to affect several important cellular processes, including cell adhesion, degradation of extracellular matrix components, and ectodomain shedding of membrane-bound growth factors that are intrinsic to cancer and various inflammatory conditions. The multiple levels by which the activity of ADAM15 can be regulated include signal transduction, modulation of catalytic function, spatial regulation, and post-translational modifications. Taken together, this multi-functional disintegrin protease not only offers a variety of potential targets for therapeutic intervention, but also represents an attractive target for pharmaceutical consideration due to its involvement in key cellular processes and various disease states. Modalities aimed at inhibiting protease activation, metalloproteinase activity, or integrin binding capability could prove beneficial for the treatment of cancer and inflammatory diseases.


Subject(s)
ADAM Proteins/drug effects , Inflammation/drug therapy , Membrane Proteins/drug effects , Neoplasms/drug therapy , Humans
19.
J Biol Chem ; 283(26): 18393-401, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18434311

ABSTRACT

The zinc-dependent disintegrin metalloproteinases (a disintegrin and metalloproteinases (ADAMs) have been implicated in several disease processes, including human cancer. Previously, we demonstrated that the expression of a catalytically active member of the ADAM family, ADAM15, is associated with the progression of prostate and breast cancer. The accumulation of the soluble ectodomain of E-cadherin in human serum has also been associated with the progression of prostate and breast cancer and is thought to be mediated by metalloproteinase shedding. Utilizing two complementary models, overexpression and stable short hairpin RNA-mediated knockdown of ADAM15 in breast cancer cells, we demonstrated that ADAM15 cleaves E-cadherin in response to growth factor deprivation. We also demonstrated that the extracellular shedding of E-cadherin was abrogated by a metalloproteinase inhibitor and through the introduction of a catalytically inactive mutation in ADAM15. We have made the novel observation that this soluble E-cadherin fragment was found in complex with the HER2 and HER3 receptors in breast cancer cells. These interactions appeared to stabilize HER2 heterodimerization with HER3 and induced receptor activation and signaling through the Erk pathway, supporting both cell migration and proliferation. In this study, we provide evidence that ADAM15 catalyzes the cleavage of E-cadherin to generate a soluble fragment that in turn binds to and stimulates ErbB receptor signaling.


Subject(s)
ADAM Proteins/chemistry , Cadherins/chemistry , Gene Expression Regulation, Neoplastic , Membrane Proteins/chemistry , Receptor, ErbB-2/metabolism , Cadherins/metabolism , Catalysis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Dimerization , Humans , Oligonucleotide Array Sequence Analysis , Protein Binding , Protein Structure, Tertiary , RNA/chemistry , Signal Transduction
20.
Cancer Res ; 68(4): 1092-9, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18281484

ABSTRACT

Using human tumor and cDNA microarray technology, we have recently shown that the ADAM15 disintegrin is significantly overexpressed during the metastatic progression of human prostate cancer. In the current study, we used lentiviral-based short hairpin RNA (shRNA) technology to down-regulate ADAM15 in the metastatic prostate cancer cell line, PC-3. ADAM15 down-regulation dramatically attenuated many of the malignant characteristics of PC-3 cells in vitro and prevented the s.c. growth of PC-3 cells in severe combined immunodeficient (SCID) mice. By inhibiting the expression of ADAM15 in PC-3 cells, we showed decreased cell migration and adhesion to specific extracellular matrix proteins. This was accompanied by a reduction in the cleavage of N-cadherin by ADAM15 at the cell surface. Fluorescence-activated cell sorting analysis revealed reduced cell surface expression of the metastasis-associated proteins alpha(v) integrin and CD44. Furthermore, matrix metalloproteinase 9 secretion and activity were abrogated in response to ADAM15 reduction. In an in vitro model of vascular invasion, loss of ADAM15 reduced PC-3 adhesion to, and migration through, vascular endothelial cell monolayers. Using an SCID mouse model of human prostate cancer metastasis, we found that the loss of ADAM15 significantly attenuated the metastatic spread of PC-3 cells to bone. Taken together, these data strongly support a functional role for ADAM15 in prostate tumor cell interaction with vascular endothelium and the metastatic progression of human prostate cancer.


Subject(s)
ADAM Proteins/physiology , Cell Communication/physiology , Endothelial Cells/pathology , Membrane Proteins/physiology , Prostatic Neoplasms/pathology , ADAM Proteins/biosynthesis , ADAM Proteins/genetics , Animals , Bone Neoplasms/secondary , Cell Adhesion/physiology , Cell Movement/physiology , Endothelial Cells/metabolism , Humans , Hyaluronan Receptors/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, SCID , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...