Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Proc Natl Acad Sci U S A ; 114(13): E2644-E2653, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28289210

ABSTRACT

Intrinsically disordered proteins (IDPs) that lack a unique 3D structure and comprise a large fraction of the human proteome play important roles in numerous cellular functions. Prostate-Associated Gene 4 (PAGE4) is an IDP that acts as a potentiator of the Activator Protein-1 (AP-1) transcription factor. Homeodomain-Interacting Protein Kinase 1 (HIPK1) phosphorylates PAGE4 at S9 and T51, but only T51 is critical for its activity. Here, we identify a second kinase, CDC-Like Kinase 2 (CLK2), which acts on PAGE4 and hyperphosphorylates it at multiple S/T residues, including S9 and T51. We demonstrate that HIPK1 is expressed in both androgen-dependent and androgen-independent prostate cancer (PCa) cells, whereas CLK2 and PAGE4 are expressed only in androgen-dependent cells. Cell-based studies indicate that PAGE4 interaction with the two kinases leads to opposing functions. HIPK1-phosphorylated PAGE4 (HIPK1-PAGE4) potentiates c-Jun, whereas CLK2-phosphorylated PAGE4 (CLK2-PAGE4) attenuates c-Jun activity. Consistent with the cellular data, biophysical measurements (small-angle X-ray scattering, single-molecule fluorescence resonance energy transfer, and NMR) indicate that HIPK1-PAGE4 exhibits a relatively compact conformational ensemble that binds AP-1, whereas CLK2-PAGE4 is more expanded and resembles a random coil with diminished affinity for AP-1. Taken together, the results suggest that the phosphorylation-induced conformational dynamics of PAGE4 may play a role in modulating changes between PCa cell phenotypes. A mathematical model based on our experimental data demonstrates how differential phosphorylation of PAGE4 can lead to transitions between androgen-dependent and androgen-independent phenotypes by altering the AP-1/androgen receptor regulatory circuit in PCa cells.


Subject(s)
Intrinsically Disordered Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Humans , Intrinsically Disordered Proteins/chemistry , Models, Molecular , Phenotype , Phosphorylation , Protein Conformation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proteome
2.
J Cell Biochem ; 118(9): 2559-2570, 2017 09.
Article in English | MEDLINE | ID: mdl-28266048

ABSTRACT

More than 90% of cancer-related deaths are caused by metastasis. Epithelial-to-Mesenchymal Transition (EMT) causes tumor cell dissemination while the reverse process, Mesenchymal-to-Epithelial Transition (MET) allows cancer cells to grow and establish a potentially deadly metastatic lesion. Recent evidence indicates that in addition to E and M, cells can adopt a stable hybrid Epithelial/Mesenchymal (E/M) state where they can move collectively leading to clusters of Circulating Tumor Cells-the "bad actors" of metastasis. EMT is postulated to occur in all four major histological breast cancer subtypes. Here, we identify a set of genes strongly correlated with CDH1 in 877 cancer cell lines, and differentially expressed genes in cell lines overexpressing ZEB1, SNAIL, and TWIST. GRHL2 and ESRP1 appear in both these sets and also correlate with CDH1 at the protein level in 40 breast cancer specimens. Next, we find that GRHL2 and CD24 expression coincide with an epithelial character in human mammary epithelial cells. Further, we show that high GRHL2 expression is highly correlated with worse relapse-free survival in all four subtypes of breast cancer. Finally, we integrate CD24, GRHL2, and ESRP1 into a mathematical model of EMT regulation to validate the role of these players in EMT. Our data analysis and modeling results highlight the relationships among multiple crucial EMT/MET drivers including ZEB1, GRHL2, CD24, and ESRP1, particularly in basal-like breast cancers, which are most similar to triple-negative breast cancer (TNBC) and are considered the most dangerous subtype. J. Cell. Biochem. 118: 2559-2570, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA-Binding Proteins/genetics , Female , Humans , Neoplasm Proteins/genetics , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics
3.
J Biol Chem ; 290(41): 25090-102, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26242913

ABSTRACT

Prostate-associated gene 4 (PAGE4) is an intrinsically disordered cancer/testis antigen that is up-regulated in the fetal and diseased human prostate. Knocking down PAGE4 expression results in cell death, whereas its overexpression leads to a growth advantage of prostate cancer cells (Zeng, Y., He, Y., Yang, F., Mooney, S. M., Getzenberg, R. H., Orban, J., and Kulkarni, P. (2011) The cancer/testis antigen prostate-associated gene 4 (PAGE4) is a highly intrinsically disordered protein. J. Biol. Chem. 286, 13985-13994). Phosphorylation of PAGE4 at Thr-51 is critical for potentiating c-Jun transactivation, an important factor in controlling cell growth, apoptosis, and stress response. Using NMR spectroscopy, we show that the PAGE4 polypeptide chain has local and long-range conformational preferences that are perturbed by site-specific phosphorylation at Thr-51. The population of transient turn-like structures increases upon phosphorylation in an ∼20-residue acidic region centered on Thr-51. This central region therefore becomes more compact and more negatively charged, with increasing intramolecular contacts to basic sequence motifs near the N and C termini. Although flexibility is decreased in the central region of phospho-PAGE4, the polypeptide chain remains highly dynamic overall. PAGE4 utilizes a transient helical structure adjacent to the central acidic region to bind c-Jun with low affinity in vitro. The binding interaction is attenuated by phosphorylation at Thr-51, most likely because of masking the effects of the more compact phosphorylated state. Therefore, phosphorylation of PAGE4 leads to conformational shifts in the dynamic ensemble, with large functional consequences. The changes in the structural ensemble induced by posttranslational modifications are similar conceptually to the conformational switching events seen in some marginally stable ("metamorphic") folded proteins in response to mutation or environmental triggers.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Prostatic Neoplasms/pathology , Amino Acid Sequence , Cell Line, Tumor , Humans , Male , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Conformation
4.
Biochim Biophys Acta ; 1842(2): 154-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24263171

ABSTRACT

The Cancer/Testis Antigen (CTA), Prostate-associated Gene 4 (PAGE4), is a stress-response protein that is upregulated in prostate cancer (PCa) especially in precursor lesions that result from inflammatory stress. In cells under stress, translocation of PAGE4 to mitochondria increases while production of reactive oxygen species decreases. Furthermore, PAGE4 is also upregulated in human fetal prostate, underscoring its potential role in development. However, the proteins that interact with PAGE4 and the mechanisms underlying its pleiotropic functions in prostatic development and disease remain unknown. Here, we identified c-Jun as a PAGE4 interacting partner. We show that both PAGE4 and c-Jun are overexpressed in the human fetal prostate; and in cell-based assays, PAGE4 robustly potentiates c-Jun transactivation. Single-molecule Förster resonance energy transfer experiments indicate that upon binding to c-Jun, PAGE4 undergoes conformational changes. However, no interaction is observed in presence of BSA or unilamellar vesicles containing the mitochondrial inner membrane diphosphatidylglycerol lipid marker cardiolipin. Together, our data indicate that PAGE4 specifically interacts with c-Jun and that, conformational dynamics may account for its observed pleiotropic functions. To our knowledge, this is the first report demonstrating crosstalk between a CTA and a proto-oncogene. Disrupting PAGE4/c-Jun interactions using small molecules may represent a novel therapeutic strategy for PCa.


Subject(s)
Antigens, Neoplasm/metabolism , Prostate/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcriptional Activation , 3' Untranslated Regions/genetics , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Prostate/embryology , Prostate/growth & development , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Conformation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological
5.
J Cell Biochem ; 116(1): 115-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25160502

ABSTRACT

Zinc finger E-box binding homeobox 1 (ZEB1) is a transcription factor that plays a central role in the epithelial to mesenchymal transition (EMT) of cancer cell lines. Studies on its regulation have mostly focused on the negative 3'UTR binding of miR200c. Interestingly, it has been previously reported that androgen receptor (AR) regulates ZEB1 expression in breast and prostate cancers. In order to validate this, various ZEB1 promoter deletions were cloned into a luciferase reporter system to elucidate the contribution of two putative androgen response elements (AREs). The in vivo contribution of AR was also assessed in cell lines after R1881 treatment using qPCR with prostate specific antigen (PSA) as the positive control. We discovered that AR upregulates the levels of expression of ZEB1 10-fold on a luciferase promoter that only contains the distal ARE. However, when the proximal ARE is included, no additional activation is apparent with AR or its hormone independent variant, AR-V7. Furthermore, we demonstrate here that a promoter construct containing both AREs activates transcription of ZEB1 even in the AR-null cell lines DU145 and PC3. Incubation of the AR-positive cell line, LNCaP with R1881, failed to substantially increase the expression levels of ZEB1. Despite the presence of AREs in the promoter region, it appears that ZEB1 expression can be induced even without AR. In addition, the region around the distal ARE is a potent repressor in AR-null cell lines.


Subject(s)
Homeodomain Proteins/metabolism , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Male , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1
6.
Biochemistry ; 53(10): 1670-9, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24559171

ABSTRACT

Prostate-associated gene 4 (PAGE4) is a cancer/testis antigen that is typically restricted to the testicular germ cells but is aberrantly expressed in cancer. Furthermore, PAGE4 is developmentally regulated with dynamic expression patterns in the developing prostate and is also a stress-response protein that is upregulated in response to cellular stress. PAGE4 interacts with c-Jun, which is activated by the stress-response kinase JNK1, and plays an important role in the development and pathology of the prostate gland. Here, we have identified homeodomain-interacting protein kinase 1 (HIPK1), also a component of the stress-response pathway, as a kinase that phosphorylates PAGE4 at T51. We show that phosphorylation of PAGE4 is critical for its transcriptional activity since mutating this T residue abolishes its ability to potentiate c-Jun transactivation. In vitro single molecule FRET indicates phosphorylation results in compaction of (still) intrinsically disordered PAGE4. Interestingly, however, while our previous observations indicated that the wild-type nonphosphorylated PAGE4 protein interacted with c-Jun [Rajagopalan , K. et al. ( 2014 ) Biochim, Biophys. Acta 1842 , 154 -163], here we show that phosphorylation of PAGE4 weakens its interaction with c-Jun in vitro. These data suggest that phosphorylation induces conformational changes in natively disordered PAGE4 resulting in its decreased affinity for c-Jun to promote interaction of c-Jun with another, unidentified, partner. Alternatively, phosphorylated PAGE4 may induce transcription of a novel partner, which then potentiates c-Jun transactivation. Regardless, the present results clearly implicate PAGE4 as a component of the stress-response pathway and uncover a novel link between components of this pathway and prostatic development and disease.


Subject(s)
Antigens, Neoplasm/metabolism , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-jun/genetics , Transcriptional Activation , Amino Acid Motifs , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Cell Line, Tumor , Humans , Male , Phosphorylation , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/physiopathology , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Stress, Physiological , Testis/metabolism
7.
J Cell Biochem ; 115(9): 1478-85, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24700698

ABSTRACT

Cancer cells can be described as an invasive species that is able to establish itself in a new environment. The concept of niche construction can be utilized to describe the process by which cancer cells terraform their environment, thereby engineering an ecosystem that promotes the genetic fitness of the species. Ecological dispersion theory can then be utilized to describe and model the steps and barriers involved in a successful diaspora as the cancer cells leave the original host organ and migrate to new host organs to successfully establish a new metastatic community. These ecological concepts can be further utilized to define new diagnostic and therapeutic areas for lethal cancers.


Subject(s)
Neoplasms/genetics , Neoplasms/pathology , Cell Survival , Ecosystem , Genetic Heterogeneity , Genomic Instability , Humans , Neoplasm Invasiveness , Tumor Microenvironment
8.
J Cell Biochem ; 115(9): 1505-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24604720

ABSTRACT

Prostate cancer is a heterogeneous disease and thus, it is important to understand whether among the heterogeneous collection of cell types, androgen-deprivation insensitive cells exist prior to hormonal manipulation. We established several LNCaP subclones with distinct insensitivities to androgen deprivation from a parental LNCaP cell line. In the resulting clones, the sensitivity to androgen-deprivation negatively correlated with their PSA expression levels. In two of these clones, an androgen insensitive clone, LNCaP-cl1, and an androgen sensitive clone, LNCaP-cl5, the DNA copy number differed significantly, indicating that these clones contain genetically distinct cells. LNCaP-cl1 had higher PSA expression but lower invasiveness and tumor growth potential than LNCaP-cl5. The expression levels of two genes that are known to be regulated by miR-21, an androgen-regulated microRNA, Sprouty1 (SPRY1) and Jagged1 (JAG1) were significantly lower in LNCaP-cl1 than in LNCaP-cl5. Knocking down SPRY1 in LNCaP cells enhanced PSA expression and cell proliferation. JAG1 administration in LNCaP cells enhanced cell invasion and JAG1 knockdown in PC3 cells suppressed cell invasion and tumor formation. These results indicated that the expression differences in SPRY1 and JAG1 may contribute to the phenotypic differences between the LNCaP-cl1 and LNCaP-cl5 clones. In tissue samples, SPRY1 expression levels were significantly lower in prostate cancer patients with PSA recurrence after surgical treatment (P = 0.0076) and JAG1 expression levels were significantly higher in Gleason sum (GS) 8-9 disease than in GS 5-6 (P = 0.0121). In summary a random population of LNCaP cells comprises a heterogeneous group of cells with different androgen-deprivation sensitivities and potential for invasiveness.


Subject(s)
Androgens/metabolism , Calcium-Binding Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Phosphoproteins/genetics , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Copy Number Variations , Gene Knockdown Techniques , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Male , Membrane Proteins/metabolism , Mice, SCID , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphoproteins/metabolism , Prognosis , Prostatic Neoplasms/surgery , Serrate-Jagged Proteins
9.
J Cell Biochem ; 114(6): 1286-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23192682

ABSTRACT

Drug resistance is a major limitation to the successful treatment of advanced prostate cancer (PCa). Patients who have metastatic, castration-resistant PCa (mCRPC) are treated with chemotherapeutics. However, these standard therapy modalities culminate in the development of resistance. We established paclitaxel resistance in a classic, androgen-insensitive mCRPC cell line (DU145) and, using a suite of molecular and biophysical methods, characterized the structural and functional changes in vitro and in vivo that are associated with the development of drug resistance. After acquiring paclitaxel-resistance, cells exhibited an abnormal nuclear morphology with extensive chromosomal content, an increase in stiffness, and faster cytoskeletal remodeling dynamics. Compared with the parental DU145, paclitaxel-resistant (DU145-TxR) cells became highly invasive and motile in vitro, exercised greater cell traction forces, and formed larger and rapidly growing tumors in mouse xenografts. Furthermore, DU145-TxR cells showed a discrete loss of keratins but a distinct gain of ZEB1, Vimentin and Snail, suggesting an epithelial-to-mesenchymal transition. These findings demonstrate, for the first time, that paclitaxel resistance in PCa is associated with a trans-differentiation of epithelial cell machinery that enables more aggressive and invasive phenotype and portend new strategies for developing novel biomarkers and effective treatment modalities for PCa patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor/drug effects , Cell Movement , Cell Nucleus/drug effects , Epithelial-Mesenchymal Transition , Humans , Inhibitory Concentration 50 , Keratin-18/metabolism , Keratin-19/metabolism , Keratin-8/metabolism , Male , Mice , Neoplasm Invasiveness , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
J Biol Chem ; 286(16): 13985-94, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21357425

ABSTRACT

The cancer/testis antigens (CTAs) are an important group of heterogeneous proteins that are predominantly expressed in the testis in the normal human adult but are aberrantly expressed in several types of cancers. Prostate-associated gene 4 (PAGE4) is a member of the CT-X family of CTAs that in addition to testis, is highly expressed in the fetal prostate, and may also play an important role both in benign and malignant prostate diseases. However, the function of this gene remains poorly understood. Here, we show that PAGE4 is a highly (100%) intrinsically disordered protein (IDP). The primary protein sequence conforms to the features of a typical IDP sequence and the secondary structure prediction algorithm metaPrDOS strongly supported this prediction. Furthermore, SDS-gel electrophoresis and analytical size exclusion chromatography of the recombinant protein revealed an anomalous behavior characteristic of IDPs. UV circular dichroism (CD) and NMR spectroscopy confirmed that PAGE4 is indeed a highly disordered protein. In further bioinformatic analysis, the PredictNLS algorithm uncovered a potential nuclear localization signal, whereas the algorithm DBS-Pred returned a 99.1% probability that PAGE4 is a DNA-binding protein. Consistent with this prediction, biochemical experiments showed that PAGE4 preferentially binds a GC-rich sequence. Silencing PAGE4 expression induced cell death via apoptosis and in mice carrying PCa xenografts, siRNA-mediated knockdown of the PAGE4 mRNA attenuated tumor growth in vivo. Furthermore, overexpressing PAGE4 protected cells from stress-induced death. To our knowledge, PAGE4 is the first example of a CTA that is an IDP with an anti-apoptotic function.


Subject(s)
Antigens, Neoplasm/chemistry , Animals , Antigens, Neoplasm/physiology , Apoptosis , Cell Line, Tumor , Circular Dichroism , Humans , Male , Mice , Mice, SCID , Neoplasm Transplantation , Prostatic Neoplasms/pathology , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Testis/metabolism
11.
Prostate ; 72(9): 966-76, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22025384

ABSTRACT

BACKGROUND: Cysteine-rich angiogenic inducer 61 (Cyr61) is an extracellular matrix protein involved in the transduction of growth factor and hormone signaling. Previously, we demonstrated that Cyr61 was highly expressed in prostate cancer (PCa) but that the expression levels were associated with a lower risk of PCa recurrence. In the present study, we demonstrate that serum Cyr61 is a potential biomarker that correlates with PCa aggressiveness. Furthermore, we also explore the potential mechanism underlying the changes in Cyr61 expression during PCa progression. METHODS: Cyr61 concentrations in the medium from PCa cell lines and in serum samples obtained from PCa patients were measured by sandwich ELISA. Serum Cyr61 levels were correlated with disease characteristics and the association between Cyr61 expression changes by several types of stimulation or stress and cAMP/cAMP-dependent protein kinase (PKA) pathway were examined. RESULTS: There was a positive correlation between Cyr61 levels in cell supernatants and mRNA expression in these cell lines. Serum Cyr61 levels were significantly higher in non-organ-confined PCa patients (116.3 ± 140.2 ng/ml) than in organ-confined PCa patients (79.7 ± 56.1 ng/ml) (P = 0.031). Cyr61 expression was up-regulated in response to both lysophosphatidic acid and androgen treatments which promoted PCa cell invasion. Serum starvation and phosphoinositide-3-kinase inhibition also resulted in Cyr61 up-regulation; however, they suppressed cell proliferation. Cyr61 up-regulation was correlated with an increase in cAMP and suppressed by PKA inhibition. CONCLUSIONS: These findings suggest that Cyr61 expression in PCa is regulated by the cAMP/PKA pathway and that circulating Cyr61 levels are a potential serum-based biomarker for characterizing PCa.


Subject(s)
Biomarkers, Tumor/blood , Cyclic AMP-Dependent Protein Kinases/physiology , Cysteine-Rich Protein 61/blood , Neoplasm Invasiveness/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Aged , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/blood , Humans , Male , Middle Aged , Prostatic Neoplasms/blood
12.
J Biol Chem ; 285(40): 30443-52, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20663877

ABSTRACT

Here, we demonstrate that p68 (DDX5) and p72 (DDX17), two homologous RNA helicases and transcriptional cofactors, are substrates for the acetyltransferase p300 in vitro and in vivo. Mutation of acetylation sites affected the binding of p68/p72 to histone deacetylases, but not to p300 or estrogen receptor. Acetylation additionally increased the stability of p68 and p72 RNA helicase and stimulated their ability to coactivate the estrogen receptor, thereby potentially contributing to its aberrant activation in breast tumors. Also, acetylation of p72, but not of p68 RNA helicase, enhanced p53-dependent activation of the MDM2 promoter, pointing at another mechanism of how p72 acetylation may facilitate carcinogenesis by boosting the negative p53-MDM2 feedback loop. Furthermore, blocking p72 acetylation caused cell cycle arrest and apoptosis, revealing an essential role for p72 acetylation. In conclusion, our report has identified for the first time that acetylation modulates RNA helicases and provides multiple mechanisms how acetylation of p68 and p72 may affect normal and tumor cells.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , DEAD-box RNA Helicases/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DEAD-box RNA Helicases/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , HeLa Cells , Humans , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors/genetics
13.
J Cell Biochem ; 112(11): 3256-67, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21748782

ABSTRACT

The cancer/testis antigens (CTAs) are a group of heterogeneous proteins that are typically expressed in the testis but aberrantly expressed in several types of cancer. Although overexpression of CTAs is frequently associated with advanced disease and poorer prognosis, the significance of this correlation is unclear since the functions of the CTAs in the disease process remain poorly understood. Here, employing a bioinformatics approach, we show that a majority of CTAs are intrinsically disordered proteins (IDPs). IDPs are proteins that, under physiological conditions in vitro, lack rigid 3D structures either along their entire length or in localized regions. Despite the lack of structure, most IDPs can transition from disorder to order upon binding to biological targets and often promote highly promiscuous interactions. IDPs play important roles in transcriptional regulation and signaling via regulatory protein networks and are often associated with dosage sensitivity. Consistent with these observations, we find that several CTAs can bind DNA, and their forced expression appears to increase cell growth implying a potential dosage-sensitive function. Furthermore, the CTAs appear to occupy "hub" positions in protein regulatory networks that typically adopt a "scale-free" power law distribution. Taken together, our data provide a novel perspective on the CTAs implicating them in processing and transducing information in altered physiological states in a dosage-sensitive manner. Identifying the CTAs that occupy hub positions in protein regulatory networks would allow a better understanding of their functions as well as the development of novel therapeutics to treat cancer.


Subject(s)
Antigens, Neoplasm/metabolism , Testis/immunology , Acetylation , Antigens, Neoplasm/genetics , Humans , Male , Phosphorylation , RNA, Messenger/genetics , SUMO-1 Protein/metabolism
14.
J Cell Biochem ; 112(4): 1066-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21308735

ABSTRACT

Creatine kinase brain (CKB) is one of three cytosolic isoforms of creatine kinase that is predominantly expressed in the brain. The enzyme is overexpressed in a wide variety of cancers, with the exception of colon cancer, where it is downregulated. The significance of this downregulation remains poorly understood. Here, we demonstrate that overexpression of CKB-C283S, a dominant-negative construct that lacks the kinase function but retains its ability to dimerize, causes remarkable changes in cell shape, adhesion, and invasion. Furthermore, it results in increased expression of stromal cell markers such as PAGE4 and SNAIL, suggesting an epithelial-to-mesenchymal transition (EMT) in these cells. In cells transfected with a CKB-expressing construct, CKB localizes not only to the cytosol but also to the nucleus, indicating a structural or kinase role unrelated to ATP storage. Furthermore, overexpression of CFP-tagged wild-type (WT) CKB in Caco-2 colon cancer cells dramatically increased the number of cells in G2/M but had little effect on cell proliferation. Taken together, these data demonstrate that the downregulation of CKB may play an important role in colon cancer progression by promoting EMT.


Subject(s)
Creatine Kinase, BB Form/genetics , Creatine Kinase, BB Form/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Caco-2 Cells , Cell Cycle , Cell Movement , Cell Nucleus/metabolism , Cell Survival , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Epithelial-Mesenchymal Transition/genetics , HCT116 Cells , Humans , Immunoblotting , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Snail Family Transcription Factors , Stress, Physiological , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/metabolism
15.
J Cell Biochem ; 112(8): 2125-37, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21465536

ABSTRACT

The microenvironment is central to many aspects of cancer pathobiology and has been proposed to play a role in the development of cancer cell resistance to therapy. To examine the response to microenvironmental conditions, two paclitaxel resistant prostate cancer (PCa) cell lines (stable and reversible) and one reversible heat resistant cell line were studied. In comparison to their parental cell lines, both paclitaxel resistant cell lines (stable and reversible) were more sensitive to microenvironmental heat, potentially yielding a synergistic therapeutic opportunity. In the two phenotypic cells repopulated after acute heat or paclitaxel treatments, there was an inverse correlation between paclitaxel and heat resistance: resistance to paclitaxel imparted sensitivity to heat; resistance to heat imparted sensitivity to paclitaxel. These studies indicate that as cancer cells evolve resistance to single microenvironmental stress they may be more sensitive to others, perhaps allowing us to design new approaches for PCa therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Hot Temperature , Hyperthermia, Induced , Paclitaxel/pharmacology , Prostatic Neoplasms/therapy , Stress, Physiological , Cell Line, Tumor , Combined Modality Therapy , Humans , Male
16.
Biochemistry ; 49(1): 1-10, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-19995069

ABSTRACT

The p68 (DDX5) and p72 (DDX17) proteins are members of the DEAD-box (DDX) family of RNA helicases. We show that both p68 and p72 are overexpressed in breast tumors. Bioinformatical analysis revealed that the SUMO pathway is upregulated in breast tumors and that both p68 and p72 contain one consensus sumoylation site, implicating that sumoylation of p68 and p72 increases during breast tumorigenesis and potentially contributes to their overexpression. We determined that p68 and p72 are indeed sumoylated at a single, homologous site. Importantly, sumoylation significantly increased the stability of p68 and p72. In contrast to p72 and consistent with an approximately 3-fold lesser half-life, p68 was found to be polyubiquitylated, and mutation of the sumoylation site increased polyubiquitylation, suggesting that sumoylation increases p68 half-life by reducing proteasomal degradation. Moreover, whereas p68 robustly coactivated transcription from an estrogen response element, its sumoylation mutant showed a drastically reduced coactivation potential. In contrast, the p68 sumoylation status did not affect the ability to enhance p53-mediated MDM2 transcription. On the contrary, preventing sumoylation of p72 caused an increase in its ability to transactivate both estrogen receptor and p53. Furthermore, sumoylation promoted the interaction of p68 and p72 with histone deacetylase 1 but had no effect on binding to histone deacetylases 2 and 3, the coactivator p300, or estrogen receptor and also did not affect homo/heterodimerization of p68/p72. In conclusion, sumoylation exerts pleiotropic effects on p68/p72, which may have important implications in breast cancer by modulating estrogen receptor and p53 activity.


Subject(s)
DEAD-box RNA Helicases/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcriptional Activation/genetics , Binding Sites , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , DEAD-box RNA Helicases/genetics , Female , Humans , Protein Stability , Receptors, Estrogen/metabolism , Transfection , Tumor Suppressor Protein p53/metabolism
17.
Biomolecules ; 9(2)2019 02 22.
Article in English | MEDLINE | ID: mdl-30813315

ABSTRACT

Folded proteins show a high degree of structural order and undergo (fairly constrained) collective motions related to their functions. On the other hand, intrinsically disordered proteins (IDPs), while lacking a well-defined three-dimensional structure, do exhibit some structural and dynamical ordering, but are less constrained in their motions than folded proteins. The larger structural plasticity of IDPs emphasizes the importance of entropically driven motions. Many IDPs undergo function-related disorder-to-order transitions driven by their interaction with specific binding partners. As experimental techniques become more sensitive and become better integrated with computational simulations, we are beginning to see how the modest structural ordering and large amplitude collective motions of IDPs endow them with an ability to mediate multiple interactions with different partners in the cell. To illustrate these points, here, we use Prostate-associated gene 4 (PAGE4), an IDP implicated in prostate cancer (PCa) as an example. We first review our previous efforts using molecular dynamics simulations based on atomistic AWSEM to study the conformational dynamics of PAGE4 and how its motions change in its different physiologically relevant phosphorylated forms. Our simulations quantitatively reproduced experimental observations and revealed how structural and dynamical ordering are encoded in the sequence of PAGE4 and can be modulated by different extents of phosphorylation by the kinases HIPK1 and CLK2. This ordering is reflected in changing populations of certain secondary structural elements as well as in the regularity of its collective motions. These ordered features are directly correlated with the functional interactions of WT-PAGE4, HIPK1-PAGE4 and CLK2-PAGE4 with the AP-1 signaling axis. These interactions give rise to repeated transitions between (high HIPK1-PAGE4, low CLK2-PAGE4) and (low HIPK1-PAGE4, high CLK2-PAGE4) cell phenotypes, which possess differing sensitivities to the standard PCa therapies, such as androgen deprivation therapy (ADT). We argue that, although the structural plasticity of an IDP is important in promoting promiscuous interactions, the modulation of the structural ordering is important for sculpting its interactions so as to rewire with agility biomolecular interaction networks with significant functional consequences.


Subject(s)
Antigens, Neoplasm/chemistry , Intrinsically Disordered Proteins/chemistry , Molecular Dynamics Simulation , Antigens, Neoplasm/metabolism , Humans , Intrinsically Disordered Proteins/metabolism , Protein Conformation
18.
Asian J Androl ; 18(5): 704-10, 2016.
Article in English | MEDLINE | ID: mdl-27427552

ABSTRACT

A striking characteristic of cancer cells is their remarkable phenotypic plasticity, which is the ability to switch states or phenotypes in response to environmental fluctuations. Phenotypic changes such as a partial or complete epithelial to mesenchymal transition (EMT) that play important roles in their survival and proliferation, and development of resistance to therapeutic treatments, are widely believed to arise due to somatic mutations in the genome. However, there is a growing concern that such a deterministic view is not entirely consistent with multiple lines of evidence, which indicate that stochasticity may also play an important role in driving phenotypic plasticity. Here, we discuss how stochasticity in protein interaction networks (PINs) may play a key role in determining phenotypic plasticity in prostate cancer (PCa). Specifically, we point out that the key players driving transitions among different phenotypes (epithelial, mesenchymal, and hybrid epithelial/mesenchymal), including ZEB1, SNAI1, OVOL1, and OVOL2, are intrinsically disordered proteins (IDPs) and discuss how plasticity at the molecular level may contribute to stochasticity in phenotypic switching by rewiring PINs. We conclude by suggesting that targeting IDPs implicated in EMT in PCa may be a new strategy to gain additional insights and develop novel treatments for this disease, which is the most common form of cancer in adult men.


Subject(s)
Intrinsically Disordered Proteins/metabolism , Prostatic Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Humans , Male , Phenotype
19.
Oncotarget ; 7(19): 27067-84, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27008704

ABSTRACT

Epithelial-to-Mesenchymal Transition (EMT) and its reverse - Mesenchymal to Epithelial Transition (MET) - are hallmarks of cellular plasticity during embryonic development and cancer metastasis. During EMT, epithelial cells lose cell-cell adhesion and gain migratory and invasive traits either partially or completely, leading to a hybrid epithelial/mesenchymal (hybrid E/M) or a mesenchymal phenotype respectively. Mesenchymal cells move individually, but hybrid E/M cells migrate collectively as observed during gastrulation, wound healing, and the formation of tumor clusters detected as Circulating Tumor Cells (CTCs). Typically, the hybrid E/M phenotype has largely been tacitly assumed to be transient and 'metastable'. Here, we identify certain 'phenotypic stability factors' (PSFs) such as GRHL2 that couple to the core EMT decision-making circuit (miR-200/ZEB) and stabilize hybrid E/M phenotype. Further, we show that H1975 lung cancer cells can display a stable hybrid E/M phenotype and migrate collectively, a behavior that is impaired by knockdown of GRHL2 and another previously identified PSF - OVOL. In addition, our computational model predicts that GRHL2 can also associate hybrid E/M phenotype with high tumor-initiating potential, a prediction strengthened by the observation that the higher levels of these PSFs may be predictive of poor patient outcome. Finally, based on these specific examples, we deduce certain network motifs that can stabilize the hybrid E/M phenotype. Our results suggest that partial EMT, i.e. a hybrid E/M phenotype, need not be 'metastable', and strengthen the emerging notion that partial EMT, but not necessarily a complete EMT, is associated with aggressive tumor progression.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells/metabolism , Cell Line, Tumor , Cell Movement/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Phenotype , RNA Interference , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
20.
Oncotarget ; 6(17): 15436-48, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-25944618

ABSTRACT

Metastasis involves multiple cycles of Epithelial-to-Mesenchymal Transition (EMT) and its reverse-MET. Cells can also undergo partial transitions to attain a hybrid epithelial/mesenchymal (E/M) phenotype that has maximum cellular plasticity and allows migration of Circulating Tumor Cells (CTCs) as a cluster. Hence, deciphering the molecular players helping to maintain the hybrid E/M phenotype may inform anti-metastasis strategies. Here, we devised a mechanism-based mathematical model to couple the transcription factor OVOL with the core EMT regulatory network miR-200/ZEB that acts as a three-way switch between the E, E/M and M phenotypes. We show that OVOL can modulate cellular plasticity in multiple ways - restricting EMT, driving MET, expanding the existence of the hybrid E/M phenotype and turning both EMT and MET into two-step processes. Our theoretical framework explains the differences between the observed effects of OVOL in breast and prostate cancer, and provides a platform for investigating additional signals during metastasis.


Subject(s)
DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/physiology , Homeodomain Proteins/genetics , MicroRNAs/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , Humans , Male , Models, Theoretical , Neoplasm Metastasis/pathology , Systems Biology/methods , Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL