Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Commun ; 13(1): 3684, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35760800

ABSTRACT

Osteolytic destruction is a hallmark of multiple myeloma, resulting from activation of osteoclast-mediated bone resorption and reduction of osteoblast-mediated bone formation. However, the molecular mechanisms underlying the differentiation and activity of osteoclasts and osteoblasts within a myelomatous microenvironment remain unclear. Here, we demonstrate that the osteocyte-expressed major histocompatibility complex class II transactivator (CIITA) contributes to myeloma-induced bone lesions. CIITA upregulates the secretion of osteolytic cytokines from osteocytes through acetylation at histone 3 lysine 14 in the promoter of TNFSF11 (encoding RANKL) and SOST (encoding sclerostin), leading to enhanced osteoclastogenesis and decreased osteoblastogenesis. In turn, myeloma cell-secreted 2-deoxy-D-ribose, the product of thymidine catalyzed by the function of thymidine phosphorylase, upregulates CIITA expression in osteocytes through the STAT1/IRF1 signaling pathway. Our work thus broadens the understanding of myeloma-induced osteolysis and indicates a potential strategy for disrupting tumor-osteocyte interaction to prevent or treat patients with myeloma bone disease.


Subject(s)
Multiple Myeloma , Osteolysis , Humans , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Nuclear Proteins , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteocytes/metabolism , Osteolysis/metabolism , Osteolysis/pathology , Osteolysis/prevention & control , RANK Ligand/metabolism , Trans-Activators , Tumor Microenvironment
2.
Cancers (Basel) ; 13(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638434

ABSTRACT

BRAF-activating mutations are the most frequent driver mutations in papillary thyroid cancer (PTC). Targeted inhibitors such as dabrafenib have been used in advanced BRAF-mutated PTC; however, acquired resistance to the drug is common and little is known about other effectors that may play integral roles in this resistance. In addition, the induction of PTC dedifferentiation into highly aggressive KRAS-driven anaplastic thyroid cancer (ATC) has been reported. We detected a novel RAC1 (P34R) mutation acquired during dabrafenib treatment in a progressive metastatic lesion with ATC phenotype. To identify a potential functional link between this novel mutation and tumor dedifferentiation, we developed a cell line derived from the metastatic lesion and compared its behavior to isogenic cell lines and primary tumor samples. Our data demonstrated that RAC1 mutations induce changes in cell morphology, reorganization of F-actin almost exclusively at the cell cortex, and changes in cell adhesion properties. We also established that RAC1 amplification, with or without mutation, is sufficient to drive cell proliferation and resistance to BRAF inhibition. Further, we identified polyploidy of chromosome 7, which harbors RAC1, in both the metastatic lesion and its derived cell line. Copy number amplification and overexpression of other genes located on this chromosome, such as TWIST1, EGFR, and MET were also detected, which might also lead to dabrafenib resistance. Our study suggests that polyploidy leading to increased expression of specific genes, particularly those located on chromosome 7, should be considered when analyzing aggressive thyroid tumor samples and in further treatments.

3.
Am J Cancer Res ; 11(6): 3227-3239, 2021.
Article in English | MEDLINE | ID: mdl-34249457

ABSTRACT

Activating transcription factor 4 (ATF4) is a crucial mediator of the integrated stress response and a negative regulator of RET tyrosine kinase receptor in medullary thyroid carcinoma (MTC). However, the impact of genomic abnormalities in the ATF4 locus on MTC pathogenesis and response to tyrosine kinase inhibitor therapy remains unknown. Here, we evaluated ATF4 copy number variation and protein levels, with overall survival and response to TKIs in a clinical cohort of fifty-nine sporadic primary MTC. We assessed the somatic RETM918T mutation by sequencing, ATF4 copy number by a real-time polymerase chain reaction, and ATF4 protein levels using immunohistochemistry. This MTC cohort comprised 45 (76%) stage IV patients with a median follow-up of 100 months (interquartile range: 58-134 months). Somatic RETM918T was present in 23/57 (40%) tumors. Mono-allelic (36%; 21/59) and bi-allelic (5%; 3/59) loss of ATF4 was identified and was associated with low ATF4 protein expression (0-20%). Kaplan-Meier curves highlight low ATF4 protein or ATF4 loss alone had a significant negative impact on median survival compared to high protein expression (P<0.001) or diploid ATF4 (P=0.011), respectively. The combination of somatic RETM918T and low ATF4 protein levels further decreased overall survival. Both allelic loss and protein reduction were associated with worse overall survival (HR=3.79, 4.06 +RETM918T , and HR=10.64, 11.66 +RETM918T , respectively). Additionally, all 4 of the 11 patients treated with TKIs with a progressive disease by RECIST had low tumor ATF4 protein, with the two partial responder's tumors having high ATF4 protein. These findings suggest that ATF4 may predict response to tyrosine kinase inhibitors, serve as a prognostic marker for personalized care, and a therapeutic target in MTC.

4.
Mol Cancer Ther ; 20(4): 665-675, 2021 04.
Article in English | MEDLINE | ID: mdl-33536187

ABSTRACT

Gain-of-function point mutations in the receptor tyrosine kinase RET, a driver oncogene in medullary thyroid carcinoma (MTC), prevent apoptosis through inhibition of ATF4, a critical transcriptional regulator of endoplasmic reticulum stress. However, the critical regulatory mechanisms driving RET-dependent oncogenesis remain elusive, and there is a clinical need to identify a transcriptional RET inhibitor. Here, we found that RET depletion decreased IGFBP2 and VEGFR2 mRNA and protein expression in MTC cells. IGFBP2 knockdown decreased cell survival and migration of MTC cells. In patients, IGFBP2 expression increased in metastatic MTC, and high IGFBP2 associated with poor overall survival. VEGFR2 protein levels were positively associated with RET expression in primary tumors, and VEGF-mediated increased cell viability was RET dependent. The small-molecule ONC201 treatment of MTC cells caused apoptotic cell death, decreased transcription of RET, VEGFR2, IGFBP2, increased mRNA levels of ATF4, and ATF4 target genes including DDIT3, BBC3, DUSP8, MKNK2, KLF9, LZTFL1, and SESN2 Moreover, IGFBP2 depletion increased ONC201-induced cell death. ONC201 inhibited tumor growth at a well-tolerated dose of 120 mg/kg/week administered by oral gavage and decreased MTC xenograft cell proliferation and angiogenesis. The protein levels of RET, IGFBP2, and VEGFR2 were decreased in ONC201-treated xenografts. Our study uncovered a novel ONC201 mechanism of action through regulation of RET and its targets, VEGFR2 and IGFBP2; this mechanism could be translated into the clinic and represent a promising strategy for the treatment of all patients with MTC, including those with TKI-refractory disease and other cancer with RET abnormalities.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Imidazoles/therapeutic use , Insulin-Like Growth Factor Binding Protein 2/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Thyroid Neoplasms/drug therapy , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Imidazoles/pharmacology , Male , Mice , Mice, Inbred NOD , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Retrospective Studies
5.
Endocr Relat Cancer ; 27(8): T27-T39, 2020 08.
Article in English | MEDLINE | ID: mdl-32580150

ABSTRACT

The 16th International Multiple Endocrine Neoplasia Workshop (MEN2019) held in Houston, TX, USA, focused on emerging topics in the pathogenesis and therapy of malignant endocrine tumors associated with MEN syndromes. With MEN-2 syndromes, the most common malignancy is medullary thyroid carcinoma (MTC). In the spirit of the original MEN meeting workshop model, the conference included didactic lectures and interactive working groups of clinicians and researchers focused on the state of science in MTC and ongoing challenges or unmet needs in the understanding of MTC and to develop strategies to address these issues.


Subject(s)
Carcinoma, Neuroendocrine/etiology , Multiple Endocrine Neoplasia/complications , Thyroid Neoplasms/etiology , Carcinoma, Neuroendocrine/pathology , Humans , Thyroid Neoplasms/pathology
6.
Oncogene ; 38(25): 5038-5049, 2019 06.
Article in English | MEDLINE | ID: mdl-30858546

ABSTRACT

Epigenetic changes that cause dysregulated gene expression during progression of androgen-independent prostate cancer (PCa) and metastatic skeletal lesions remain elusive. Here, we explored the role of histone demethylase NO66 in the pathogenesis of PCa and bone metastasis-related skeletal lesions. Tissue and cDNA microarrays of PCa were analyzed for NO66 mRNA and protein levels. We examined the effects of gain and loss of NO66 function on cell viability, colony formation, migration, invasion, and tumor-induced skeletal lesions in femoral bone. RNAseq and ChIPseq were performed to elucidate NO66-target genes in PCa. We report that NO66 levels were upregulated in advanced primary prostate tumors compared to normal tissue or tumors with low Gleason scores. Forced expression of NO66 promoted cell survival and invasion of PCa cells; whereas, knockdown of NO66 resulted in decreased cell survival and increased sensitivity to docetaxel. NO66-overexpressing PC3 cells implanted into the femoral bone of male SCID mice caused massive bone loss and stimulation of mouse osteoclast-promoting genes, including Dickkopf1, Cathepsin K, Nf-kß,; and Calcr, suggesting a role for NO66 in tumor growth in bone and osteoclast activity. Combined RNAseq and ChIP-seq revealed that NO66 activates the survival gene MCL1, the invasion-associated genes IGFBP5 and MMP3, the pro-oncogenic genes CTNNB1 and CCND1, and the epigenetic modifier gene KMT2A in androgen-independent PCa. Our findings uncover the role of NO66 as a key oncogenic driver in PCa, causing osteolytic lesions through upstream epigenetic regulation of key genes for survival, invasion and metastasis, and pro-osteoclastic factors.


Subject(s)
Cell Transformation, Neoplastic/genetics , Dioxygenases/physiology , Histone Demethylases/physiology , Osteolysis/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Dioxygenases/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone Demethylases/genetics , Histones/metabolism , Humans , Male , Mice , Mice, SCID , NIH 3T3 Cells , Osteolysis/pathology , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/metabolism
7.
Mol Cancer Res ; 17(3): 751-760, 2019 03.
Article in English | MEDLINE | ID: mdl-30552230

ABSTRACT

Medullary thyroid carcinoma (MTC) originates from the C cells of the thyroid gland, which secrete calcitonin. Lymph node and distant metastases are frequently present at diagnosis. Activating mutations of RET, a driver oncogene in MTC that encodes a tyrosine kinase receptor, prevents apoptosis through inhibition of ATF4, a key transcriptional regulator of endoplasmic reticulum (ER) stress. We hypothesized that the combination of a tyrosine kinase inhibitor (TKI) and an ATF4 inducer promotes cell death by triggering catastrophic oxidative stress and apoptotic cell death. Here, we report that the ER-associated protein degradation (ERAD) inhibitor eeyarestatin sensitized MTC cells to the TKIs, sunitinib and vandetanib, thereby leading to synergistic upregulation of ATF4 expression, accumulation of reactive oxygen species, and subsequent cell death. Genome-wide analysis of ATF4 interaction sites by chromatin immunoprecipitation (ChIP) sequencing revealed that among ATF4 target genes was KLF9 (Kruppel-like factor 9), which induces MTC apoptosis. ChIP assays revealed that ATF4 occupancy at the KLF9 promoter was increased in MTC cells treated with eeyarestatin or vandetanib alone and was further enhanced in cells treated with both drugs, leading to increased KLF9 transcription. Depletion of ATF4 by shRNA led to downregulation of KLF9 expression and prevented oxidative stress-induced cell death. Furthermore, we identified ATF4 target genes (LZTFL1, MKNK2, and SIAH1 with known tumor suppressor function) that were synergistically upregulated with the combination of TKI and ERAD inhibitor. IMPLICATIONS: These findings reveal a combination therapy that induces reactive oxygen species-dependent catastrophic cell death through induction of ATF4 and KLF9 transcriptional activity.


Subject(s)
Activating Transcription Factor 4/genetics , Apoptosis/drug effects , Endoplasmic Reticulum-Associated Degradation/drug effects , Kruppel-Like Transcription Factors/genetics , Oxidative Stress/drug effects , Protein Kinase Inhibitors/therapeutic use , Activating Transcription Factor 4/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Protein Kinase Inhibitors/pharmacology
8.
J Clin Endocrinol Metab ; 102(3): 933-941, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27935748

ABSTRACT

Context: Medullary thyroid cancer (MTC) is an aggressive tumor that harbors activating mutations of the RET proto-oncogene. We previously reported that RET inhibits transcriptional activity of ATF4, the master regulator of the stress response pathway, to prevent cell death. Objective: We hypothesized that loss of function of ATF4 plays a role in initiation of MTC. Design: Targeted deletion of Atf4 in mice was used to assess ATF4 function in the thyroid gland. ATF4 overexpression was achieved by adenoviral and lentiviral vectors. We used immunohistochemical analysis and western blotting of MTC tumors to determine protein levels of RET and ATF4 and the Kaplan-Meier method to determine their association with clinical outcome. Results: Targeted deletion of Atf4 in mice causes C-cell hyperplasia, a precancerous lesion for MTC. Forced ATF4 expression decreased survival of MTC cells and blocked the activation of RET downstream signaling pathways (phosphorylated ERK, phosphorylated AKT, and p70S6K). ATF4 knockdown decreased sensitivity to tyrosine kinase inhibitor-induced apoptosis. Moreover, ATF4 expression decreased RET protein levels by promoting RET ubiquitination. We found decreased or loss of ATF4 in 52% of MTC tumors (n = 39) compared with normal thyroid follicle cells. A negative correlation was observed between RET and ATF4 protein levels in MTC tumors, and low ATF4 expression was associated with poor overall survival in patients with MTC. Conclusions: ATF4 was identified as a negative regulator of RET, a candidate tumor suppressor gene, and may be a molecular marker that distinguishes patients at high risk of MTC from those with a longer survival prognosis.


Subject(s)
Activating Transcription Factor 4/genetics , Apoptosis/genetics , Carcinoma, Neuroendocrine/genetics , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Activating Transcription Factor 4/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Anilides/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Neuroendocrine/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Female , Genes, Tumor Suppressor , HEK293 Cells , Humans , Immunohistochemistry , Indoles/pharmacology , Kaplan-Meier Estimate , Male , Mice , Mice, Knockout , Middle Aged , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Pyridines/pharmacology , Pyrroles/pharmacology , Real-Time Polymerase Chain Reaction , Sunitinib , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Neoplasms/metabolism , Ubiquitination/drug effects , Ubiquitination/genetics , Young Adult
9.
J Biol Chem ; 290(18): 11749-61, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25795775

ABSTRACT

The RET proto-oncogene, a tyrosine kinase receptor, is widely known for its essential role in cell survival. Germ line missense mutations, which give rise to constitutively active oncogenic RET, were found to cause multiple endocrine neoplasia type 2, a dominant inherited cancer syndrome that affects neuroendocrine organs. However, the mechanisms by which RET promotes cell survival and prevents cell death remain elusive. We demonstrate that in addition to cytoplasmic localization, RET is localized in the nucleus and functions as a tyrosine-threonine dual specificity kinase. Knockdown of RET by shRNA in medullary thyroid cancer-derived cells stimulated expression of activating transcription factor 4 (ATF4), a master transcription factor for stress-induced apoptosis, through activation of its target proapoptotic genes NOXA and PUMA. RET knockdown also increased sensitivity to cisplatin-induced apoptosis. We observed that RET physically interacted with and phosphorylated ATF4 at tyrosine and threonine residues. Indeed, RET kinase activity was required to inhibit the ATF4-dependent activation of the NOXA gene because the site-specific substitution mutations that block threonine phosphorylation increased ATF4 stability and activated its targets NOXA and PUMA. Moreover, chromatin immunoprecipitation assays revealed that ATF4 occupancy increased at the NOXA promoter in TT cells treated with tyrosine kinase inhibitors or the ATF4 inducer eeyarestatin as well as in RET-depleted TT cells. Together these findings reveal RET as a novel dual kinase with nuclear localization and provide mechanisms by which RET represses the proapoptotic genes through direct interaction with and phosphorylation-dependent inactivation of ATF4 during the pathogenesis of medullary thyroid cancer.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Proto-Oncogene Proteins c-ret/metabolism , Activating Transcription Factor 4/chemistry , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cisplatin/pharmacology , Gene Expression Regulation/drug effects , Humans , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Threonine/metabolism , Transcription, Genetic/drug effects
10.
Carcinogenesis ; 34(10): 2244-52, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23722650

ABSTRACT

Chemotherapeutic agents have been the mainstay of cancer therapy for years. However, their effectiveness has been limited by toxicities they impart on normal cells. Staurosporine (ST) has been shown to arrest normal, but not breast cancer, cells in G1. Therefore, ST may become a chemoprotective agent, arresting normal cells while allowing tumor cells to enter cell cycle phases where they are sensitive to chemotherapeutic agents. Understanding the mechanism of ST-mediated G1 arrest may allow for a beneficial chemoprotective treatment strategy for patients. We utilized 76NE6 (pRb+/p53-), 76NF2V (pRb+/p53+) and 76NE7 (pRb-/P53+) non-tumorigenic human mammary epithelial cell lines to understand the role of the Rb and p53 pathways in ST-directed G1 arrest. CDK4 was downregulated by ST in Rb+ cells, but its presence could not reverse the arrest, neither did its stable downregulation alter ST-mediated cellular response. ST-mediated G1 arrest required pRb, which in turn initiated a cascade of events leading to inhibition of CDK4. Further assessment of this pathway revealed that Chk1 expression and activity were required for the Rb-dependent arrest. For example, pRb+ cells with small interfering RNA to Chk1 had approximately 60% less cells in G1 phase compared with controls and pRb- cells do not arrest upon ST. Furthermore, Chk1 expression facilitates the release of the Rb+ cells from G1 arrest. Collectively, our data suggest that pRb cooperates with Chk1 to mediate a G1 arrest only in pRb+ cells. The elucidation of this pathway can help identify novel agents to protect cancer patients against the debilitating effects of chemotherapy.


Subject(s)
Enzyme Inhibitors/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/physiology , Protective Agents/pharmacology , Protein Kinases/metabolism , Retinoblastoma Protein/metabolism , Staurosporine/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Transformed , Checkpoint Kinase 1 , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Gene Expression , Humans , Protein Kinases/genetics , Retinoblastoma Protein/genetics , Signal Transduction/drug effects
11.
J Cell Biol ; 190(1): 101-14, 2010 Jul 12.
Article in English | MEDLINE | ID: mdl-20603326

ABSTRACT

Here we provide evidence in support of an inherent role for Arpc1b, a component of the Arp2/3 complex, in regulation of mitosis and demonstrate that its depletion inhibits Aurora A activation at the centrosome and impairs the ability of mammalian cells to enter mitosis. We discovered that Arpc1b colocalizes with gamma-tubulin at centrosomes and stimulates Aurora A activity. Aurora A phosphorylates Arpc1b on threonine 21, and expression of Arpc1b but not a nonphosphorylatable Arpc1b mutant in mammalian cells leads to Aurora A kinase activation and abnormal centrosome amplification in a Pak1-independent manner. Together, these findings reveal a new function for Arpc1b in centrosomal homeostasis. Arpc1b is both a physiological activator and substrate of Aurora A kinase and these interactions help to maintain mitotic integrity in mammalian cells.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Centrosome/metabolism , Enzyme Activators/metabolism , Mitosis/physiology , Protein Serine-Threonine Kinases/metabolism , Actin-Related Protein 2-3 Complex/genetics , Aurora Kinases , Cell Line, Tumor , Humans , Mutation , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Tubulin/genetics , Tubulin/metabolism
12.
Cancer Res ; 70(12): 5085-95, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20530684

ABSTRACT

The cyclin E-cyclin-dependent kinase 2 (CDK2) complex accelerates entry into the S phase of the cell cycle and promotes polyploidy, which may contribute to genomic instability in cancer cells. The effect of low molecular weight isoforms of cyclin E (LMW-E) overexpression on mitotic progression and its link to genomic instability were the focus of this study. Here, we show that full-length cyclin E (EL) and LMW-E overexpression impairs the G(2)-M transition differently by targeting dual-specificity phosphatase Cdc25C activity. We identify Cdc25C as an interaction partner and substrate for cyclin E/CDK2 kinase. Specifically, the cyclin E/CDK2 complex phosphorylates Cdc25C on Ser(214), leading to its premature activation, which coincides with higher cyclin B/CDK1 and Polo-like kinase 1 (PLK1) activities in an S-phase-enriched population that result in faster mitotic entry. Whereas EL overexpression leads to hyperactivation of Cdc25C, cyclin B/CDK1, and PLK1 in a G(2)-M-enriched population, LMW-E overexpression causes premature inactivation of Cdc25C and PLK1, leading to faster mitotic exit. In addition, LMW-E-overexpressing cells showed a reduction in the mitotic index in the presence of a spindle poison and faster degradation of cyclin B, suggesting an increased rate of mitotic slippage and adaptation to the spindle checkpoint. Lastly, downregulation of Cdc25C inhibits LMW-E-mediated chromosome missegregation, anaphase bridges, and centrosome amplification. These results suggest that the high levels of LMW-E isoforms found in breast cancer may contribute to cellular transformation and genomic instability by impairing mitotic progression involving Cdc25C.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Division/physiology , Cyclin E/metabolism , Mitosis/genetics , Oncogene Proteins/metabolism , S Phase/physiology , cdc25 Phosphatases/metabolism , Blotting, Western , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Centrosome/pathology , Chromosomal Instability , Cyclin B/metabolism , Cyclin E/genetics , Cyclin-Dependent Kinase 2/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Molecular Weight , Oncogene Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/pharmacology , cdc25 Phosphatases/antagonists & inhibitors , cdc25 Phosphatases/genetics , Polo-Like Kinase 1
13.
Cancer Res ; 70(12): 5074-84, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20530685

ABSTRACT

Overexpression of the low molecular weight isoforms (LMW-E) of cyclin E induces chromosome instability; however, the degree to which these tumor-specific forms cause genomic instability differs from that of full-length cyclin E (EL), and the underlying mechanism(s) has yet to be elucidated. Here, we show that EL and LMW-E overexpression impairs the G(2)-M transition differently and leads to different degrees of chromosome instability in a breast cancer model system. First, the most significant difference is that EL overexpression prolongs cell cycle arrest in prometaphase, whereas LMW-E overexpression reduces the length of mitosis and accelerates mitotic exit. Second, LMW-E-overexpressing cells are binucleated or multinucleated with amplified centrosomes, whereas EL-overexpressing cells have the normal complement of centrosomes. Third, LMW-E overexpression causes mitotic defects, chromosome missegregation during metaphase, and anaphase bridges during anaphase, most of which are not detected on EL induction. LMW-E induces additional mitotic defects in cooperation with p53 loss in both normal and tumor cells. Fourth, LMW-E-overexpressing cells fail to arrest in the presence of nocodazole. Collectively, the mitotic defects mediated by LMW-E induction led to failed cytokinesis and polyploidy, suggesting that LMW-E expression primes cells to accrue chromosomal instability by shortening the length of mitosis. Lastly, LMW-E expression in human breast cancer tissues correlates with centrosome amplification and higher nuclear grade. These results suggest that LMW-E overexpression leads to higher centrosome numbers in breast cancer, which is a prerequisite for genomic instability.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Centrosome/pathology , Chromosomal Instability , Cyclin E/metabolism , Mitosis/genetics , Oncogene Proteins/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cell Line, Tumor , Cyclin E/genetics , Cytoplasm/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Middle Aged , Molecular Weight , Oncogene Proteins/genetics , Ploidies
14.
Cancer Res ; 67(15): 7062-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671172

ABSTRACT

Metastasis-associated protein 1 (MTA1), a component of the nuclear remodeling complex and the founding homologue of the MTA family, has been implicated in metastasis, but definitive causative evidence in an animal model system is currently lacking. Here, we show that MTA1 overexpression in transgenic mice is accompanied by a high incidence of spontaneous B cell lymphomas including diffuse large B cell lymphomas (DLBCL). Lymphocytes and lymphoma cells from MTA1-TG mice are hyperproliferative. Lymphomas were transplantable and of clonal origin and were characterized by down-regulation of p27Kip1 as well as up-regulation of Bcl2 and cyclin D1. The significance of these murine studies was established by evidence showing a widespread up-regulation of MTA1 in DLBCL from humans. These findings reveal a previously unrecognized role for the MTA1 pathway in the development of spontaneous B cell lymphomas, and offer a potential therapeutic target in B cell lymphomas. These observations suggest that MTA1-TG mice represent a new model of spontaneous DLBCL associated with high tumor incidence and could be used for therapeutic intervention studies.


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Neoplastic/physiology , Lymphoma, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Transcription Factors/genetics , Animals , Blotting, Southern , Cell Proliferation , Female , Histone Deacetylases/genetics , Humans , Lymph Nodes/pathology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/etiology , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Tumor Cells, Cultured
15.
Cancer Res ; 67(15): 7132-8, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671180

ABSTRACT

Previously, we have shown that metastasis-associated protein 1 (MTA1) overexpression in transgenic mice was accompanied by high incidence of spontaneous B-cell lymphomas including diffuse large B-cell lymphomas (DLBCL). To understand the molecular basis of lymphoma in MTA1-transgenic (MTA1-TG) mice, we wished to identify a putative MTA1 target with a causal role in B-cell lymphogenesis. Using chromatin immunoprecipitation assays, we identified paired box gene 5 (Pax5), a molecule previously implicated in B-cell lymphogenesis, as a potential downstream effector of MTA1. Lymphomas from MTA1-TG mice also showed up-regulation of Pax5. We also found that MTA1 acetylated on Lys(626) interacted with p300 histone acetyltransferase, and that acetylated MTA1 was recruited to the Pax5 promoter to stimulate Pax5 transcription. Global gene profiling identified down-regulation of a set of genes, including those downstream of Pax5 and directly implicated in the B-cell lymphogenesis. Significance of these murine studies was established by evidence showing a widespread up-regulation of both MTA1 and Pax5 in DLBCL from humans. These observations provide in vivo genetic evidence for a role of MTA1 in lymphomagenesis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Lymphoma, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , PAX5 Transcription Factor/genetics , Transcription Factors/physiology , Animals , Blotting, Northern , Chromatin Immunoprecipitation , Gene Expression Profiling , Histone Deacetylase 1 , Histone Deacetylases/genetics , Humans , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Plasmids , Promoter Regions, Genetic , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Transcriptional Activation , Transfection , Tumor Cells, Cultured
16.
Int J Cancer ; 118(11): 2703-10, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16381000

ABSTRACT

Mammalian LIM kinase 1 (LIMK1) phosphorylates and inactivates the actin-binding and -depolymerizing factor cofilin and induces actin cytoskeletal changes. LIMK1 is reported to play an important role in cell motility, but the mechanism of induction of cell motility and the role of LIMK1 in tumor growth, angiogenesis and invasion are poorly understood. Here we show that expression of LIMK1 in MDA-MB-435 human breast cancer cells enhanced cell proliferation and cell invasiveness and promoted in vitro angiogenesis. Since tumor metastasis requires degradation of the extracellular matrix by the serine protease urokinase type plasminogen activator (uPA), we examined the role of LIMK1 in the regulation of uPA/uPAR system. LIMK1 overexpression in breast cancer cells upregulated the uPA system, increased uPA promoter activity, induced uPA and uPAR mRNA and protein expression and induced uPA secretion. In contrast, cells transfected with the catalytically inactive LIMK mutant D460N-LIMK1 did not exhibit these phenotypic changes. Blocking antibodies against uPA and uPAR suppressed LIMK1-induced cell invasiveness. In addition, LIMK1 overexpression increased tumor growth in female athymic nude mice, promoted tumor angiogenesis and induced metastasis to livers and lungs, possibly by increasing uPA expression in the tumors. Finally, LIMK1 and uPAR were coordinately overexpressed in human breast tumors. These results suggested an important role for LIMK1 signaling in breast cancer tumor growth, angiogenesis and invasion and a regulatory connection between LIMK1 and the uPA system.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/physiology , Neoplasm Metastasis/physiopathology , Protein Kinases/biosynthesis , Protein Kinases/metabolism , Animals , Extracellular Matrix/metabolism , Female , Gene Expression Profiling , Humans , Lim Kinases , Mice , Mice, Nude , Neovascularization, Pathologic , Phosphorylation , Promoter Regions, Genetic , Signal Transduction , Transfection , Tumor Cells, Cultured , Up-Regulation
17.
Development ; 131(14): 3469-79, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15226262

ABSTRACT

Emerging data suggest that metastasis-associated protein 1 (MTA1) represses ligand-dependent transactivation functions of estrogen receptor-alpha in cultured breast cancer cells and that MTA1 is upregulated in human breast tumors. However, the role of MTA1 in tumorigenesis in a physiologically relevant animal system remains unknown. To reveal the role of MTA1 in mammary gland development, transgenic mice expressing MTA1 under the control of the mouse mammary tumor virus promoter long terminal repeat were generated. Unexpectedly, we found that mammary glands of these virgin transgenic mice exhibited extensive side branching and precocious differentiation because of increased proliferation of ductal and alveolar epithelial cells. Mammary glands of virgin transgenic mice resemble those from wild-type mice in mid-pregnancy and inappropriately express beta-casein, cyclin D1 and beta-catenin protein. Increased ductal growth was also observed in the glands of ovariectomized female mice, as well as of transgenic male mice. MTA1 dysregulation in mammary epithelium and cancer cells triggered downregulation of the progesterone receptor-B isoform and upregulation of the progesterone receptor-A isoform, resulting in an imbalance in the native ratio of progesterone receptor A and B isoforms. MTA1 transgene also increased the expression of progesterone receptor-A target genes Bcl-XL (Bcl2l1) and cyclin D1 in mammary gland of virgin mice, and, subsequently, produced a delayed involution. Remarkably, 30% of MTA1 transgenic females developed focal hyperplastic nodules, and about 7% exhibited mammary tumors within 18 months. These studies establish, for the first time, a potential role of MTA1 in mammary gland development and tumorigenesis. The underlying mechanism involves the upregulation of progesterone receptor A and its targets, Bcl-XL and cyclin D1.


Subject(s)
Gene Expression Regulation, Developmental , Histone Deacetylases/physiology , Mammary Glands, Animal/embryology , Repressor Proteins/physiology , Animals , Blotting, Northern , Blotting, Southern , Bromodeoxyuridine/pharmacology , Cell Division , Cyclin D1/metabolism , Female , Immunoblotting , In Situ Nick-End Labeling , Male , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Transgenic , Models, Genetic , Protein Isoforms , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trans-Activators , bcl-X Protein
18.
Cancer Cell ; 5(6): 575-85, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15193260

ABSTRACT

We identified dynein light chain 1 (DLC1) as a physiologic substrate of p21-activated kinase 1 (Pak1). Pak1-DLC1 interaction plays an essential role in cell survival, which depends on Pak1's phosphorylation of DLC1 on Ser88. Pak1 associates with the complex of DLC1 and BimL, a proapoptotic BH3-only protein, and phosphorylates both proteins. Phosphorylation of BimL by Pak1 prevents it from interacting with and inactivation of Bcl-2, an antiapoptotic protein. Overexpression of DLC1 but not DLC1-Ser88Ala mutant promotes cancerous properties of breast cancer cells. DLC1 protein level is elevated in more than 90% of human breast tumors. The regulation of cell survival functions by Pak1-DLC1 interaction represents a novel mechanism by which a signaling kinase might regulate the cancerous phenotypes.


Subject(s)
Apoptosis , Carrier Proteins/pharmacology , Drosophila Proteins , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Alanine/chemistry , Blotting, Western , Breast Neoplasms/pathology , Cell Cycle , Cell Division , Cell Line, Tumor , Cell Survival , Cell Transformation, Neoplastic , Dyneins , Flow Cytometry , Gene Expression Regulation, Neoplastic , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , Microscopy, Fluorescence , Mutation , Phenotype , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Serine/chemistry , Signal Transduction , Time Factors , Two-Hybrid System Techniques , Up-Regulation , p21-Activated Kinases
19.
Clin Cancer Res ; 10(2): 658-67, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14760089

ABSTRACT

PURPOSE: Abnormalities in the expression and signaling pathways downstream of the epidermal growth factor receptor (EGFR) contribute to the progression, invasion, and maintenance of the malignant phenotype in human cancers, including those of the head and neck and breast. Accordingly, agents such as the EGFR tyrosine kinase inhibitor (EGFR-TKI) ZD1839 (Iressa) are promising, biologically based treatments that are in various stages of preclinical and clinical development. The process of tumor progression requires, among other steps, increased transformation, directional migration, and enhanced cell survival; this study explored the effect of ZD1839 on the stimulation of c-Src and p21-activated kinase 1 (Pak1), which are vital for transformation, directional motility, and cell survival of cancer cells. EXPERIMENTAL DESIGN: We examined the effect of ZD1839 on biochemical and functional assays indicative of directional motility and cell survival, using human head and neck squamous cancer cells and breast cancer cells. RESULTS: ZD1839 effectively inhibited c-Src activation and Pak1 activity in exponentially growing cancer cells. In addition, ZD1839 suppressed EGF-induced stimulation of EGFR autophosphorylation on Y1086 and Grb2-binding Y1068 sites, c-Src phosphorylation on Y215, and Pak1 activity. ZD1839 also blocked EGF-induced cytoskeleton remodeling, redistribution of activated EGFR, and in vitro invasiveness of cancer cells. CONCLUSIONS: These studies suggest that the EGFR-TKI ZD1839 may cause potent inhibition of the Pak1 and c-Src pathways and, therefore, have potential to affect the invasiveness of human cancer cells deregulated in these growth factor receptor pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Quinazolines/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , Cell Survival , Cytoskeleton/metabolism , Disease Progression , Dose-Response Relationship, Drug , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Gefitinib , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Microscopy, Fluorescence , Neoplasm Invasiveness , Phenotype , Phosphorylation , Precipitin Tests , Signal Transduction , Transfection , Vinculin/metabolism , p21-Activated Kinases , src-Family Kinases
20.
EMBO Rep ; 5(2): 154-60, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14749719

ABSTRACT

The formation of new branched actin filament networks at the cell cortex of migrating cells is choreographed by the actin-related protein (Arp) 2/3 complex. Despite the fundamental role of the Arp2/3 complex in actin nucleation and branching, upstream signals that control the functions of p41-Arc, a putative regulatory component of the mammalian Arp2/3 complex, remain unidentified. Here we show that p41-Arc interacts with p21-activated kinase 1 (Pak1) both in vitro and in vivo. Pak1 phosphorylation of p41-Arc regulates its localization with the Arp2/3 complex in the cortical nucleation regions of cells. Pak1 phosphorylates p41-Arc on threonine 21 in the first WD repeat, and its mutation has functional implications in vivo. Threonine 21 phosphorylation by Pak1 is required for both constitutive and growth-factor-induced cell motility. Pak1 regulation of p41-Arc activation status represents a novel mechanism by which signalling pathways may influence the functions of the Arp2/3 complex, leading to motility in mammalian cells.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Protein Serine-Threonine Kinases/metabolism , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex , Actins/chemistry , Actins/genetics , Cell Movement/drug effects , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Gene Library , Glutathione Transferase/analysis , Humans , Immunoprecipitation , Mutation/genetics , Mutation/physiology , Phalloidine/chemistry , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Threonine/genetics , Threonine/metabolism , Wound Healing/physiology , p21-Activated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...