Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cancer Res ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38959335

ABSTRACT

The TP53 tumor suppressor is frequently altered in lethal, castration-resistant prostate cancer (CRPC). However, to date there are no effective treatments that specifically target TP53 alterations. Using transcriptomic and metabolomic analyses, we showed here that TP53-altered prostate cancer (PCa) exhibits an increased dependency on asparagine and overexpresses asparagine synthetase (ASNS), the enzyme catalyzing the synthesis of asparagine. Mechanistically, loss or mutation of TP53 transcriptionally activated ASNS expression, directly as well as via mTORC1-mediated ATF4 induction, driving de novo asparagine biosynthesis to support CRPC growth. TP53-altered CRPC cells were sensitive to asparagine restriction by knockdown of ASNS or L-asparaginase treatment to deplete the intracellular and extracellular sources of asparagine, respectively, and cell viability was rescued by asparagine addition. Notably, pharmacological inhibition of intracellular asparagine biosynthesis using a glutaminase inhibitor and depletion of extracellular asparagine with L-asparaginase significantly reduced asparagine production and effectively impaired CRPC growth. This study highlights the significance of ASNS-mediated metabolic adaptation as a synthetic vulnerability in CRPC with TP53 alterations, providing a rationale for targeting asparagine production to treat these lethal prostate cancers.

2.
Cancer Res ; 82(11): 2110-2123, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35405009

ABSTRACT

Androgen receptor (AR) pathway inhibitors are the mainstay treatment for advanced prostate cancer, but resistance to therapy is common. Here, we used a CRISPR activation screen in metastatic castration-sensitive prostate cancer cells to identify genes that promote resistance to AR inhibitors. Activation of the TGFß target gene paired-related homeobox2 (PRRX2) promoted enzalutamide resistance. PRRX2 expression was the highest in double-negative prostate cancer (DNPC), which lack AR signaling and neuroendocrine differentiation, and a PRRX2-related gene signature identified a subset of patients with DNPC with reduced overall survival. PRRX2-expressing cells showed alterations in the CDK4/6/Rb/E2F and BCL2 pathways. Accordingly, treatment with CDK4/6 and BCL2 inhibitors sensitized PRRX2-expressing, castration-resistant tumors to enzalutamide. Overall, PRRX2 was identified as a driver of enzalutamide resistance. The PRRX2 signature merits investigation as a biomarker of enzalutamide resistance in prostate cancer that could be reversed with CDK4/6 and BCL2 inhibitors. SIGNIFICANCE: PRRX2 mediates enzalutamide resistance via activation of the E2F and BCL2 pathways, which can be targeted with CDK4/6 and BCL2 inhibitors to reverse resistance.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms, Castration-Resistant , Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Resistance, Neoplasm/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Nitriles/therapeutic use , Phenylthiohydantoin , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Androgen/metabolism
3.
Cancer Res ; 81(8): 2157-2170, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33637566

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer with poor prognosis, and there is a critical need for novel therapeutic approaches. NEPC is associated with molecular perturbation of several pathways, including amplification of MYCN. Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase involved in the pathogenesis of neuroblastoma and other malignancies where it cooperates with N-Myc. We previously identified the first case of ALK F1174C-activating mutation in a patient with de novo NEPC who responded to the ALK inhibitor, alectinib. Here, we show that coactivation of ALK and N-Myc (ALK F1174C/N-Myc) is sufficient to transform mouse prostate basal stem cells into aggressive prostate cancer with neuroendocrine differentiation in a tissue recombination model. A novel gene signature from the ALK F1174C/N-Myc tumors was associated with poor outcome in multiple human prostate cancer datasets. ALK F1174C and ALK F1174C/N-Myc tumors displayed activation of the Wnt/ß-catenin signaling pathway. Chemical and genetic ALK inhibition suppressed Wnt/ß-catenin signaling and tumor growth in vitro in NEPC and neuroblastoma cells. ALK inhibition cooperated with Wnt inhibition to suppress NEPC and neuroblastoma proliferation in vitro and tumor growth and metastasis in vivo. These findings point to a role for ALK signaling in NEPC and the potential of cotargeting the ALK and Wnt/ß-catenin pathways in ALK-driven tumors. Activated ALK and N-Myc are well known drivers in neuroblastoma development, suggesting potential similarities and opportunities to elucidate mechanisms and therapeutic targets in NEPC and vice versa. SIGNIFICANCE: These findings demonstrate that coactivation of ALK and N-Myc induces NEPC by stimulating the Wnt/ß-catenin pathway, which can be targeted therapeutically.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Carcinoma, Neuroendocrine/etiology , N-Myc Proto-Oncogene Protein/metabolism , Prostatic Neoplasms/etiology , Wnt Signaling Pathway , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Animals , Carbazoles/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Humans , Male , Mice , Mutation , N-Myc Proto-Oncogene Protein/genetics , Neoplastic Stem Cells , Neuroblastoma/drug therapy , Neuroblastoma/etiology , Neuroblastoma/pathology , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Exome Sequencing , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
4.
Cancer Cell ; 36(5): 483-497.e15, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31679823

ABSTRACT

Small molecules that directly target MYC and are also well tolerated in vivo will provide invaluable chemical probes and potential anti-cancer therapeutic agents. We developed a series of small-molecule MYC inhibitors that engage MYC inside cells, disrupt MYC/MAX dimers, and impair MYC-driven gene expression. The compounds enhance MYC phosphorylation on threonine-58, consequently increasing proteasome-mediated MYC degradation. The initial lead, MYC inhibitor 361 (MYCi361), suppressed in vivo tumor growth in mice, increased tumor immune cell infiltration, upregulated PD-L1 on tumors, and sensitized tumors to anti-PD1 immunotherapy. However, 361 demonstrated a narrow therapeutic index. An improved analog, MYCi975 showed better tolerability. These findings suggest the potential of small-molecule MYC inhibitors as chemical probes and possible anti-cancer therapeutic agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/pharmacology , Drug Discovery/methods , Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Feasibility Studies , Female , Humans , Male , Mice , Neoplasms/immunology , Neoplasms/pathology , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Threonine/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
5.
J Natl Cancer Inst ; 111(3): 311-321, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30312426

ABSTRACT

BACKGROUND: Recurrence following androgen-deprivation therapy is associated with adverse clinical outcomes in prostate cancer, but the cellular origins and molecular mechanisms underlying this process are poorly defined. We previously identified a population of castration-resistant luminal progenitor cells expressing Bmi1 in the normal mouse prostate that can serve as a cancer cell-of-origin. Here, we investigate the potential of Bmi1-expressing tumor cells that survive castration to initiate recurrence in vivo. METHODS: We employed lineage retracing in Bmi1-CreER; R26R-confetti; Ptenf/f transgenic mice to mark and follow the fate of emerging recurrent tumor clones after castration. A tissue recombination strategy was used to rescue transgenic mouse prostates by regeneration as grafts in immunodeficient hosts. We also used a small molecule Bmi1 inhibitor, PTC-209, to directly test the role of Bmi1 in recurrence. RESULTS: Transgenic prostate tumors (n = 17) regressed upon castration but uniformly recurred within 3 months. Residual regressed tumor lesions exhibited a transient luminal-to-basal phenotypic switch and marked cellular heterogeneity. Additionally, in these lesions, a subpopulation of Bmi1-expressing castration-resistant tumor cells overexpressed the stem cell reprogramming factor Sox2 (mean [SD] = 41.1 [3.8]%, n = 10, P < .001). Bmi1+Sox2+ cells were quiescent (BrdU+Bmi1+Sox2+ at 3.4 [1.5]% vs BrdU+Bmi1+Sox2- at 18.8 [3.4]%, n = 10, P = .009), consistent with a cancer stem cell phenotype. By lineage retracing, we established that recurrence emerges from the Bmi1+ tumor cells in regressed tumors. Furthermore, treatment with the small molecule Bmi1 inhibitor PTC-209 reduced Bmi1+Sox2+ cells (6.1 [1.4]% PTC-209 vs 38.8 [2.3]% vehicle, n = 10, P < .001) and potently suppressed recurrence (retraced clone size = 2.6 [0.5] PTC-209 vs 15.7 [5.9] vehicle, n = 12, P = .04). CONCLUSIONS: These results illustrate the utility of lineage retracing to define the cellular origins of recurrent prostate cancer and identify Bmi1+Sox2+ cells as a source of recurrence that could be targeted therapeutically.


Subject(s)
Cell Lineage , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , SOXB1 Transcription Factors/metabolism , Animals , Cell Proliferation , Humans , Male , Mice , Mice, Transgenic , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , SOXB1 Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Oncotarget ; 8(31): 51264-51276, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881646

ABSTRACT

Genomic studies are rapidly identifying genetic alterations in human cancer, but functional validation of such alterations has been slow. Here, using human prostate cancer as a model, we have assessed the feasibility of engineering defined genetic alterations in well-known cancer driver genes to transform benign prostate epithelial organoids derived from African American men. Benign human prostate organoids were transduced with lentiviruses expressing MYC, shPTEN, shTP53 and AR, alone and in various combinations, to recapitulate prostate cancer development. Organoids expressing MYC, shPTEN, shTP53 and AR (denoted MPPA); MYC, shPTEN and shTP53 (MPP); or MYC (M) were significantly larger, had higher proliferation rates and demonstrated pathologically transformed morphology compared to organoids transduced with control lentivirus. Alterations in MYC, PTEN and TP53 also affected the rate of organoid basal-to-luminal differentiation in vitro. MPPA and MPP organoids expressed the clinical prostate cancer marker AMACR and developed prostate adenocarcinoma when grafted under the renal capsule in mice. These data indicate that genetic alterations commonly observed in human prostate cancer can be rapidly modeled in human organoid culture. Prostate cancer organoids provide a useful pre-clinical model for the evaluation of new candidate cancer genes, cancer disparities, and potentially for testing of novel therapeutic agents.

7.
Nat Commun ; 7: 12943, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27703144

ABSTRACT

Identification of defined cell populations with stem/progenitor properties is key for understanding prostate development and tumorigenesis. Here we show that the polycomb repressor protein Bmi1 marks a population of castration-resistant luminal epithelial cells enriched in the mouse proximal prostate. We employ lineage tracing to show that these castration-resistant Bmi1-expressing cells (or CARBs) are capable of tissue regeneration and self-renewal. Notably, CARBs are distinct from the previously described luminal castration-resistant Nkx3.1-expressing cells (CARNs). CARBs can serve as a prostate cancer cell-of-origin upon Pten deletion, yielding luminal prostate tumours. Clonal analysis using the R26R-confetti allele indicates preferential tumour initiation from CARBs localized to the proximal prostate. These studies identify Bmi1 as a marker for a distinct population of castration-resistant luminal epithelial cells enriched in the proximal prostate that can serve as a cell of origin for prostate cancer.


Subject(s)
Polycomb Repressive Complex 1/metabolism , Prostate/cytology , Prostatic Neoplasms, Castration-Resistant/metabolism , Proto-Oncogene Proteins/metabolism , Regeneration , Alleles , Androgens/metabolism , Animals , Cell Lineage , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Male , Mice , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Recombination, Genetic
8.
World J Surg Oncol ; 12: 268, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25141859

ABSTRACT

BACKGROUND: Researchers in recent studies have reported that the sonic hedgehog (Shh) signaling pathway plays a crucial role during tumorigenesis, angiogenesis and cellular differentiation. We investigated the clinical and pathological significances of the Shh pathway and of its lymphangiogenic components in non-small-cell lung cancer (NSCLC), namely, Shh, glioma-associated oncogene homolog zinc finger protein 1 (Gli1), lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) and vascular endothelial growth factor D (VEGF-D). METHODS: The expression of Shh, Gli1, LYVE-1 and VEGF-D in primary NSCLC tissue from 40 patients was examined using immunohistochemical assays, and relationships between expression and clinicopathological data, such as age, gender, histology, tumor size, nodal stage, visceral pleural invasion, lymphatic thromboembolism, recurrence and overall survival were investigated. RESULTS: Of the 40 specimens examined, 25 (62.5%), 20 (50.0%), 11 (27.5%) and 20 (50.0%) were positive for Shh, Gli1, LYVE-1 or VEGF-D expression, respectively. The expression of Gli1 and LYVE-1 were significantly associated (P = 0.011), and Shh and LYVE-1 expression was related to visceral pleural invasion and lymphatic thromboembolism, respectively (P < 0.05). Shh expression levels compared on survival curves were statistically significant in univariate logrank analysis (P = 0.020). However, other clinicopathological factors did not reveal any statistical significance in univariate and multivariate analyses. CONCLUSIONS: To our knowledge, this the first report of the relationship between components of the Shh signaling pathway and prognosis in NSCLC. The expression of Shh, Gli1 and LYVE-1 was found to be associated with clinicopathological factors and survival. Thus, the overexpression of the Shh signaling pathway could serve as a predictor of malignant behavior, including lymphangiogenesis, in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Hedgehog Proteins/metabolism , Lung Neoplasms/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor D/metabolism , Vesicular Transport Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Adenocarcinoma, Bronchiolo-Alveolar/mortality , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Female , Follow-Up Studies , Hedgehog Proteins/genetics , Humans , Immunoenzyme Techniques , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lymphangiogenesis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate , Thromboembolism , Transcription Factors/genetics , Vascular Endothelial Growth Factor D/genetics , Vesicular Transport Proteins/genetics , Zinc Finger Protein GLI1
9.
Am J Pathol ; 183(5): 1679-1687, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24012679

ABSTRACT

Human papillomavirus is the main cause of cervical cancer, yet other nonviral cofactors are also required for the disease. The uterine cervix is a hormone-responsive tissue, and female hormones have been implicated in cervical carcinogenesis. A transgenic mouse model expressing human papillomavirus oncogenes E6 and/or E7 has proven useful to study a mechanism of hormone actions in the context of this common malignancy. Estrogen and estrogen receptor α are required for the development of cervical cancer in this mouse model. Estrogen receptor α is known to up-regulate expression of the progesterone receptor, which, on activation by its ligands, either promotes or inhibits carcinogenesis, depending on the tissue context. Here, we report that progesterone receptor inhibits cervical and vaginal epithelial cell proliferation in a ligand-dependent manner. We also report that synthetic progestin medroxyprogesterone acetate promotes regression of cancers and precancerous lesions in the female lower reproductive tracts (ie, cervix and vagina) in the human papillomavirus transgenic mouse model. Our results provide the first experimental evidence that supports the hypothesis that progesterone signaling is inhibitory for cervical carcinogenesis in vivo.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Progesterone/metabolism , Signal Transduction , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/prevention & control , Animals , Carcinoma, Squamous Cell/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cervix Uteri/metabolism , Cervix Uteri/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Estrogen Receptor alpha/metabolism , Female , Humans , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Transgenic , Mucins/metabolism , Papillomavirus E7 Proteins/metabolism , Progesterone/pharmacology , Receptors, Progesterone/deficiency , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Uterine Cervical Neoplasms/metabolism
10.
Exp Cell Res ; 318(5): 539-49, 2012 Mar 10.
Article in English | MEDLINE | ID: mdl-22227409

ABSTRACT

p53, the major human tumor suppressor, appears to be related to sonic hedgehog (Shh)-Gli-mediated tumorigenesis. However, the role of p53 in tumor progression by the Shh-Gli signaling pathway is poorly understood. Herein we investigated the critical regulation of Gli3-p53 in tumorigenesis of colon cancer cells and the molecular mechanisms underlying these effects. RT-PCR analysis indicated that the mRNA level of Shh and Gli3 in colon tumor tissues was significantly higher than corresponding normal tissues (P<0.001). The inhibition of Gli3 by treatment with Gli3 siRNA resulted in a clear decrease in cell proliferation and enhanced the level of expression of p53 proteins compared to treatment with control siRNA. The half-life of p53 was dramatically increased by treatment with Gli3 siRNA. In addition, treatment with MG132 blocked MDM2-mediated p53 ubiquitination and degradation, and led to accumulation of p53 in Gli3 siRNA-overexpressing cells. Importantly, ectopic expression of p53 siRNA reduced the ability of Gli3 siRNA to suppress proliferation of those cells compared with the cells treated with Gli3 siRNA alone. Moreover, Gli3 siRNA sensitized colon cancer cells to treatment with anti-cancer agents (5-FU and bevacizumab). Taken together, our studies demonstrate that loss of Gli3 signaling leads to disruption of the MDM2-p53 interaction and strongly potentiate p53-dependent cell growth inhibition in colon cancer cells, indicating a basis for the rational use of Gli3 antagonists as a novel treatment option for colon cancer.


Subject(s)
Gene Expression , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Bevacizumab , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms , Fluorouracil/pharmacology , Gene Knockdown Techniques , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Signal Transduction , Tumor Suppressor Protein p53/genetics , Ubiquitination , Zinc Finger Protein Gli3
11.
Cancer Res ; 71(22): 7061-70, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21975935

ABSTRACT

Activation of sonic hedgehog (Shh) signaling has been implicated in progression of a variety of tumors. In this study, we elucidated a role for Shh in the invasion of gastric tumors and determined the mechanism by which Shh is regulated. Immunohistochemical analysis of 178 primary human gastric tumor biopsies indicated that Shh expression was positively correlated with lymph node metastasis, high lymphatic vessel density, and poor prognosis. In mouse xenograft models of human gastric cancer, enforced expression of Shh significantly enhanced the incidence of lung metastasis compared with nonexpressing controls. Mechanistic investigations revealed that phosphoinositide 3-kinase (PI3K)/Akt inhibition blocked Shh-induced epithelial-mesenchyme transition, the activity of matrix metalloproteinase 9 (MMP-9), and lymphangiogenesis, reducing tumor invasiveness and metastasis. Taken together, our findings establish that Shh signaling promotes the metastasis of gastric cancer through activation of the PI3K/Akt pathway, which leads to mesenchymal transition and MMP-9 activation. These findings offer preclinical validation of Shh as a candidate therapeutic target for treatment of metastatic gastric cancers.


Subject(s)
Epithelial-Mesenchymal Transition , Hedgehog Proteins/physiology , Lymphangiogenesis , Matrix Metalloproteinase 9/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Stomach Neoplasms/pathology , Adult , Aged , Animals , Cell Line, Tumor , Female , Hedgehog Proteins/analysis , Humans , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Middle Aged , Phosphatidylinositol 3-Kinases/physiology
13.
Exp Cell Res ; 317(12): 1746-62, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21570392

ABSTRACT

Bone morphogenetic proteins (BMPs) have been implicated in tumorigenesis and metastatic progression in various types of cancer cells, but the role and cellular mechanism in the invasive phenotype of gastric cancer cells is not known. Herein, we determined the roles of phosphoinositide 3-kinase (PI3K)/AKT, extracellular signal-regulated protein kinase (ERK), nuclear factor (NF)-κB, and matrix metalloproteinase (MMP) expression in BMP-2-mediated metastatic function in gastric cancer. We found that stimulation of BMP-2 in gastric cancer cells enhanced the phosphorylation of AKT and ERK. Accompanying activation of AKT and ERK kinase, BMP-2 also enhanced phosphorylation/degradation of IκBα and the nuclear translocation/activation of NF-κB. Interestingly, blockade of PI3K/AKT and ERK signaling using LY294002 and PD98059, respectively, significantly inhibited BMP-2-induced motility and invasiveness in association with the activation of NF-κB. Furthermore, BMP-2-induced MMP-9 expression and enzymatic activity was also significantly blocked by treatment with PI3K/AKT, ERK, or NF-κB inhibitors. Immunohistochemistry staining of 178 gastric tumor biopsies indicated that expression of BMP-2 and MMP-9 had a significant positive correlation with lymph node metastasis and a poor prognosis. These results indicate that the BMP-2 signaling pathway enhances tumor metastasis in gastric cancer by sequential activation of the PI3K/AKT or MAPK pathway followed by the induction of NF-κB and MMP-9 activity, indicating that BMP-2 has the potential to be a therapeutic molecular target to decrease metastasis.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intestinal Neoplasms/secondary , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/pathology , Apoptosis , Blotting, Western , Bone Morphogenetic Protein 2/genetics , Cell Adhesion , Cell Movement , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , Immunoenzyme Techniques , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Luciferases/metabolism , Lymphatic Metastasis , Male , Matrix Metalloproteinase 9/genetics , Middle Aged , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Rate , Tumor Cells, Cultured , Wound Healing
14.
Exp Cell Res ; 316(1): 24-37, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19835871

ABSTRACT

Up-regulation of bone morphogenetic proteins (BMPs) and their receptors by tumor is an important hallmark in cancer progression, as it contributes through autocrine and paracrine mechanisms to tumor development, invasion, and metastasis. Generally, increased motility and invasion are positively correlated with the epithelial-mesenchymal transition (EMT). The purpose of the present study was to determine whether BMP-2 signaling to induce gastric cancer cells to undergo EMT-mediated invasion might pass through the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Herein we showed that gastric cancer cell lines express all the components of BMP-2 signaling, albeit to different extents. Moreover, an increased concentration of BMP-2 strongly enhanced motility and invasiveness in gastric cancer cells, whereas no increase was observed in cells treated with either Noggin (a BMP-2 inhibitor) or BMP-2 blocking antibodies. The stimulation of BMP-2 in gastric cancer cells induces a full EMT characterized by Snail induction, E-cadherin delocalization and down-regulation, and up-regulation of mesenchymal and invasiveness markers. Furthermore, blockade of BMP-2 signaling by Noggin or BMP-2 blocking antibodies also restored these changes in EMT markers. In addition, phosphorylation of Akt was also enhanced by treatment with BMP-2, but not Noggin or BMP-2 blocking antibodies. Pretreatment of gastric cancer cells with PI-3 kinase/Akt kinase inhibitor (kinase-dead Akt [DN-Akt], Akt siRNA, or LY294002) significantly inhibited BMP-2-induced EMT and invasiveness. Overall, our studies suggest that BMP-2 promotes motility and invasion of gastric cancer cells by activating PI-3 kinase/Akt and that targeting of this signaling pathway may provide therapeutic opportunities in preventing metastasis mediated by BMP-2.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Cell Movement/physiology , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Stomach Neoplasms/pathology , Antibodies/immunology , Antibodies/pharmacology , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein Receptors/genetics , Cadherins/metabolism , Carrier Proteins/pharmacology , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromones/pharmacology , Collagen , Drug Combinations , Enzyme Inhibitors , Gene Expression/genetics , Genes, Reporter/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Laminin , Morpholines/pharmacology , Nerve Tissue Proteins/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proteoglycans , RNA, Small Interfering/genetics , Smad Proteins/metabolism , Snail Family Transcription Factors , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Vimentin/metabolism
15.
Oncol Rep ; 22(3): 525-34, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639199

ABSTRACT

Although dysregulation of bone morphogenetic protein (BMP) signaling has been linked to various types of cancers, the relationship between abnormal activation of these signaling pathways and tumorigenesis is not clear. The purpose of the current study was to clarify how BMP2 is involved in colon cancer aggressiveness. The data showed that SW480 and DLD-1 cells displayed different responses to short- and long-term exposure to BMP2. During the first 24 h of exposure to BMP2, these cells were growth-inhibited, whereas surviving cells became resistant to growth inhibition, showing epithelial-to-mesenchymal transformation (EMT) and enhanced motility and invasiveness. Interestingly, in highly metastatic mesenchymal colon carcinoma cells (CT26), blockade of BMP2 signaling by BMP2 siRNA prevented EMT, motility and invasiveness; rather, blockade of BMP2 signaling caused a mesenchymal-to-epithelial transition (MET). The levels of phosphorylated Akt were very different between the two cell types; the BMP2-sensitive SW480 and DLD-1 cells had much higher levels of expression than the BMP2-resistant SW480 and DLD-1 and CT26 cells. CT26 cells, following exposure to BMP2 and activation of Akt, escaped the EMT-induced cellular motility and invasiveness. Moreover, LY294002 treatment of BMP2-sensitive SW480 cells blocked cell growth and enhanced motility and invasiveness. Together, these results suggest that suppression of the PI3 kinase/Akt pathway is correlated with the development of BMP2 resistance and invasion in BMP2-induced EMT in colon cancer.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Colonic Neoplasms/etiology , Epithelial Cells/pathology , Mesoderm/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Cell Line, Tumor , Cell Transformation, Neoplastic , Colonic Neoplasms/pathology , Humans , Neoplasm Invasiveness , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
16.
Differentiation ; 77(5): 450-61, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19375841

ABSTRACT

The interaction between mammary epithelial cells and their surrounding microenvironment are important in the development of the mammary gland. Thus, mesenchymal stem cells (MSCs), which retain pluripotency for various mesenchymal lineages, may provide a permissive environment for the morphologic alteration and differentiation of mammary epithelial cells. To this end, we investigated whether the interactions between mammary epithelial cells and human placenta-derived MSCs (hPMSC) affect the morphology, proliferation, and differentiation of epithelial cells in a co-culture system. We show that after co-culture with hPMSCs, human mammary epithelial cell lines (MCF-10F and HEMC) underwent significant morphologic alterations and a dramatic increase in ductal-alveolar branching, which was accompanied by a decrease or loss of the epithelial marker E-cadherin and a gain of the mesenchymal markers, alpha-SMA and vimentin. MCF-10F and HEMC proliferation was also inhibited in the presence of hPMSCs, and this retardation in growth was due to cell cycle arrest. Furthermore, in MCF-10F and HMEC cells, hPMSCs induced the production of lipid droplets, milk fat globule protein, and milk protein lactoferrin, which are markers of functional mammary differentiation. We also noticed an elevation in ALK5 and phosphorylated Smad3 protein levels upon hPMSC co-culture. Strikingly, the changes in morphology, proliferation, and differentiation were reversed by treatment with ALK5 or Smad3 knockdown in MCF-10F/hPMSC co-cultures. Collectively, our findings suggest that co-cultivation with hPMSCs leads to epithelial to mesenchymal transition (EMT) and differentiation of human breast epithelial cells through the ALK5/Smad3 signaling pathway.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad3 Protein/metabolism , Breast/cytology , Cell Differentiation , Coculture Techniques , Epithelium/metabolism , Female , Humans , Mesoderm/cytology , Mesoderm/metabolism , Placenta/cytology , Pregnancy , Receptor, Transforming Growth Factor-beta Type I
17.
Carcinogenesis ; 29(3): 480-90, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18174246

ABSTRACT

It is known that the activation of hedgehog (Hh) signaling is involved in the progression and invasion of various tumors, including gastric carcinoma. In this study, we investigated the impact of transforming growth factor (TGF)-beta signaling on the sonic hedgehog (Shh)-mediated invasion of gastric cancer cells. We found that higher concentrations of N-Shh enhanced cell motility and invasiveness in gastric cancer cells, whereas no increase was observed in cells that were treated with KAAD-cyclopamine (a Shh signaling inhibitor) or anti-Shh blocking antibodies. In addition, the N-Shh-induced migration and invasiveness of gastric cancer cells were reduced by treatment with anti-TGF-beta blocking antibody or TGF-beta1 small interfering RNA (siRNA) in presence of N-Shh when compared with control groups. Furthermore, TGF-beta1 secretion, TGF-beta-mediated transcriptional response, expression of activin receptor-like kinase (ALK) 5 protein and phosphorylation of Smad 3 were also enhanced by treatment with N-Shh, but not KAAD-cyclopamine, anti-Shh or TGF-beta1 blocking antibodies. Blockade of the ALK5 kinase in the presence of N-Shh significantly inhibited phosphorylation of Smad 3, activity of matrix metalloproteinases and Shh-induced cell motility/invasiveness. Importantly, transient expression of ALK5 siRNA or Smad 3 siRNA reduced the ability of N-Shh to stimulate migration and invasion of those cells compared with the cells treated with non-specific control siRNA. In summary, these results indicate that Shh promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway. Additionally, our findings are the first to suggest a role and mechanism for Shh signaling as it relates to the metastatic potential of gastric cancer, thereby indicating potential therapeutic molecular targets to decrease metastasis.


Subject(s)
Hedgehog Proteins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad3 Protein/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta/physiology , Base Sequence , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Primers , Humans , Immunohistochemistry , Lymphatic Metastasis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Phosphorylation , RNA, Small Interfering , Receptor, Transforming Growth Factor-beta Type I , Stomach Neoplasms/enzymology , Stomach Neoplasms/metabolism
18.
Biochim Biophys Acta ; 1783(3): 438-47, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18164268

ABSTRACT

Development of acquired resistance to tamoxifen is a major clinical problem during endocrine treatment in estrogen receptor positive breast cancer. Transforming growth factor-beta1 (TGF-beta) has been implicated in tamoxifen-induced cellular signaling in breast cancer, and increased Akt activation is associated with tamoxifen-resistant cell types. We hypothesized that the relationship between TGF-beta and Akt signaling may be involved in the development and progression of tamoxifen resistance. Tamoxifen-resistant (Tam-R) cells were established from parental MCF-7 cells by continuously exposing them to 4-hydroxytamoxifen (4-OHT). Tam-R cells were associated with a decrease in TGF-beta1 secretion, TGF-beta-mediated transcriptional response, and growth inhibitory effects of 4-OHT. Tam-R cells expressed significantly higher levels of phosphorylated Akt and lower levels of phosphorylated Smad 3 in both the absence and presence of 4-OHT when compared to MCF-7 cells treated with 4-OHT. Ectopic expression of constitutively active Akt (Myc-Akt(Myr)) rendered MCF-7 cells resistant to activation by TGF-beta and the growth inhibitory effects of 4-OHT, while over-expression of kinase-dead Akt (Myc-Akt(K179M)) or LY294002 treatment of Tam-R cells enhanced TGF-beta activation and blocked cell growth. These results suggest that suppression of TGF-beta signaling by activated Akt is correlated with the development of tamoxifen resistance in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-akt/physiology , Tamoxifen , Transforming Growth Factor beta/physiology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Carcinoma/drug therapy , Carcinoma/enzymology , Cell Line, Tumor , Drug Resistance, Neoplasm/physiology , Humans , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Tamoxifen/therapeutic use , Transforming Growth Factor beta/metabolism
19.
Int J Oncol ; 29(6): 1541-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17088994

ABSTRACT

The constitutive activation of JNK has been implicated in Ras-induced cellular transformation and activated JNK is down-regulated by the tumor suppressor protein, RASSF1A. In this study, we examined whether RASSF1A blocked oncogenic Ras-induced JNK activation. Exogenous expression of H-RasG12V induced JNK phosphorylation and RASSF1A co-transfected with H-RasG12V efficiently suppressed Ras-triggered JNK activation in various cancer cell lines. RASSF1A expression revived the H-RasG12V-induced p27Kip1 down-regulation. JNK siRNA treatment also promoted recovery from the H-RasG12V-induced p27Kip1 down-regulation. These results demonstrate that RASSF1A inhibited H-RasG12V-induced JNK activation and JNK-mediated p27Kip1 down-regulation. From these results, we propose that RASSF1A exerts a tumor-suppressing effect by blocking oncogenic Ras-induced JNK activation.


Subject(s)
Genes, ras , MAP Kinase Kinase 4/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enzyme Activation , Humans , MAP Kinase Kinase 4/biosynthesis , MAP Kinase Kinase 4/genetics , RNA, Small Interfering/genetics , Transfection , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , ras Proteins/biosynthesis , ras Proteins/genetics , ras Proteins/metabolism
20.
Biochem Biophys Res Commun ; 338(2): 1179-84, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16256947

ABSTRACT

Ribosomal proteins not only act as components of the translation apparatus but also regulate cell proliferation and apoptosis. A previous study reported that MRPL41 plays an important role in p53-dependent apoptosis. It also showed that MRPL41 arrests the cell cycle by stabilizing p27(Kip1) in the absence of p53. This study found that MRPL41 mediates the p21(WAF1/CIP1)-mediated G1 arrest in response to serum starvation. The cells were released from serum starvation-induced G1 arrest via the siRNA-mediated blocking of MRPL41 expression. Overall, these results suggest that MRPL41 arrests the cell cycle by increasing the p21(WAF1/CIP1) and p27(Kip1) levels under the growth inhibitory conditions.


Subject(s)
Adenocarcinoma/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase , Lung Neoplasms/metabolism , Ribosomal Proteins/metabolism , Cell Line, Tumor , Culture Media, Serum-Free , Humans , Mitochondrial Proteins , Serum/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...