Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 19(7): 1123-1136, 2021 07.
Article in English | MEDLINE | ID: mdl-33846123

ABSTRACT

Prostate cancer genomic subtypes that stratify aggressive disease and inform treatment decisions at the primary stage are currently limited. Previously, we functionally validated an aggressive subtype present in 15% of prostate cancer characterized by dual deletion of MAP3K7 and CHD1. Recent studies in the field have focused on deletion of CHD1 and its role in androgen receptor (AR) chromatin distribution and resistance to AR-targeted therapy; however, CHD1 is rarely lost without codeletion of MAP3K7. Here, we show that in the clinically relevant context of co-loss of MAP3K7 and CHD1 there are significant, collective changes to aspects of AR signaling. Although CHD1 loss mainly impacts the expansion of the AR cistrome, loss of MAP3K7 drives increased AR target gene expression. Prostate cancer cell line models engineered to cosuppress MAP3K7 and CHD1 also demonstrated increased AR-v7 expression and resistance to the AR-targeting drug enzalutamide. Furthermore, we determined that low protein expression of both genes is significantly associated with biochemical recurrence (BCR) in a clinical cohort of radical prostatectomy specimens. Low MAP3K7 expression, however, was the strongest independent predictor for risk of BCR over all other tested clinicopathologic factors including CHD1 expression. Collectively, these findings illustrate the importance of MAP3K7 loss in a molecular subtype of prostate cancer that poses challenges to conventional therapeutic approaches. IMPLICATIONS: These findings strongly implicate MAP3K7 loss as a biomarker for aggressive prostate cancer with significant risk for recurrence that poses challenges for conventional androgen receptor-targeted therapies.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , MAP Kinase Kinase Kinases/genetics , Prostatic Neoplasms/genetics , RNA Interference , Receptors, Androgen/genetics , Signal Transduction/genetics , Androgens/pharmacology , Benzamides/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Logistic Models , MAP Kinase Kinase Kinases/metabolism , Male , Neoplasm Recurrence, Local , Nitriles/pharmacology , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Risk Factors
2.
Mol Cancer Res ; 17(10): 1985-1998, 2019 10.
Article in English | MEDLINE | ID: mdl-31300540

ABSTRACT

The combined loss of CHD1 and MAP3K7 promotes aggressive prostate cancer by unknown mechanisms. Because both of these genes are lost genetically in prostate cancer, they cannot be directly targeted. We applied an established computational systems pharmacology approach (TRAP) to identify altered signaling pathways and associated druggable targets. We compared gene expression profiles of prostate cancer with coloss of CHD1 and MAP3K7 with prostate cancer diploid for these genes using The Cancer Genome Atlas patient samples. This analysis prioritized druggable target genes that included CDK1 and CDK2. We validated that inhibitors of these druggable target genes, including the CDK1/CDK2 inhibitor dinaciclib, had antiproliferative and cytotoxic effects selectively on mouse prostate cells with knockdown of Chd1 and Map3k7. Dinaciclib had stronger effects on prostate cells with suppression of Map3k7 independent of Chd1 and also compared with cells without loss of Map3k7. Dinaciclib treatment reduced expression of homologous recombination (HR) repair genes such as ATM, ATR, BRCA2, and RAD51, blocked BRCA1 phosphorylation, reduced RAD51 foci formation, and increased γH2AX foci selectively in prostate cells with suppression of Map3k7, thus inhibiting HR repair of chromosomal double-strand breaks. Dinaciclib-induced HR disruption was also observed in human prostate cells with knockdown of MAP3K7. Cotreatment of dinaciclib with DNA-damaging agents or PARP inhibitor resulted in a stronger cytotoxic effect on prostate cells with suppression of MAP3K7 compared with those without loss of MAP3K7, or to each single agent. IMPLICATIONS: These findings demonstrate that loss of MAP3K7 is a main contributing factor to drug response through disruption of HR in prostate cancer.


Subject(s)
DNA Damage/drug effects , Homologous Recombination/genetics , MAP Kinase Kinase Kinases/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/pathology
3.
Elife ; 42015 Oct 27.
Article in English | MEDLINE | ID: mdl-26506153

ABSTRACT

Prostate cancers with mutations to a protein called SPOP use an error-prone method to repair broken DNA strands.


Subject(s)
Genomic Instability , Nuclear Proteins/deficiency , Prostatic Neoplasms/pathology , Repressor Proteins/deficiency , Animals , Humans , Male
4.
Cancer Res ; 75(6): 1021-34, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25770290

ABSTRACT

Prostate cancer subtypes are poorly defined and functional validation of drivers of ETS rearrangement-negative prostate cancer has not been conducted. Here, we identified an ETS(-) subtype of aggressive prostate cancer (ERG(-)MAP3K7(del)CHD1(del)) and used a novel developmental model and a cell line xenograft model to show that cosuppression of MAP3K7 and CHD1 expression promotes aggressive disease. Analyses of publicly available prostate cancer datasets revealed that MAP3K7 and CHD1 were significantly codeleted in 10% to 20% of localized tumors and combined loss correlated with poor disease-free survival. To evaluate the functional impact of dual MAP3K7-CHD1 loss, we suppressed Map3k7 and/or Chd1 expression in mouse prostate epithelial progenitor/stem cells (PrP/SC) and performed tissue recombination experiments in vivo. Dual shMap3k7-shChd1 PrP/SC recombinants displayed massive glandular atypia with regions of prostatic intraepithelial neoplasia and carcinoma apparent. Combined Map3k7-Chd1 suppression greatly disrupted normal prostatic lineage differentiation; dual recombinants displayed significant androgen receptor loss, increased neuroendocrine differentiation, and increased neural differentiation. Clinical samples with dual MAP3K7-CHD1 loss also displayed neuroendocrine and neural characteristics. In addition, dual Map3k7-Chd1 suppression promoted E-cadherin loss and mucin production in recombinants. MAP3K7 and CHD1 protein loss also correlated with Gleason grade and E-cadherin loss in clinical samples. To further validate the phenotype observed in the PrP/SC model, we suppressed MAP3K7 and/or CHD1 expression in LNCaP prostate cancer cells. Dual shMAP3K7-shCHD1 LNCaP xenografts displayed increased tumor growth and decreased survival compared with shControl, shMAP3K7, and shCHD1 xenografts. Collectively, these data identify coordinate loss of MAP3K7 and CHD1 as a unique driver of aggressive prostate cancer development.


Subject(s)
DNA Helicases/physiology , DNA-Binding Proteins/physiology , MAP Kinase Kinase Kinases/physiology , Prostatic Neoplasms/pathology , Animals , Cadherins/analysis , Cell Line, Tumor , Cells, Cultured , Disease Progression , Humans , Male , Mice , Neoplasm Grading , Neoplasm Invasiveness
5.
Mol Cancer Ther ; 13(10): 2361-71, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25122071

ABSTRACT

Prostate cancer is the most commonly diagnosed malignancy among Western men and accounts for the second leading cause of cancer-related deaths. Prostate cancer tends to grow slowly and recent studies suggest that it relies on lipid fuel more than on aerobic glycolysis. However, the biochemical mechanisms governing the relationships between lipid synthesis, lipid utilization, and cancer growth remain unknown. To address the role of lipid metabolism in prostate cancer, we have used etomoxir and orlistat, clinically safe drugs that block lipid oxidation and lipid synthesis/lipolysis, respectively. Etomoxir is an irreversible inhibitor of the carnitine palmitoyltransferase (CPT1) enzyme that decreases ß oxidation in the mitochondria. Combinatorial treatments using etomoxir and orlistat resulted in synergistic decreased viability in LNCaP, VCaP, and patient-derived benign and prostate cancer cells. These effects were associated with decreased androgen receptor expression, decreased mTOR signaling, and increased caspase-3 activation. Knockdown of CPT1A enzyme in LNCaP cells resulted in decreased palmitate oxidation but increased sensitivity to etomoxir, with inactivation of AKT kinase and activation of caspase-3. Systemic treatment with etomoxir in nude mice resulted in decreased xenograft growth over 21 days, underscoring the therapeutic potential of blocking lipid catabolism to decrease prostate cancer tumor growth.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Epoxy Compounds/pharmacology , Lactones/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Epoxy Compounds/administration & dosage , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Lactones/administration & dosage , Lipid Metabolism/drug effects , Male , Metabolism , Mice , Mice, Nude , Orlistat , Oxidation-Reduction/drug effects , Prostatic Neoplasms/pathology , Random Allocation , Signal Transduction , Xenograft Model Antitumor Assays
6.
Endocr Relat Cancer ; 21(4): T147-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24872510

ABSTRACT

Androgen receptor (AR) signaling is vital to the development and function of the prostate and is a key pathway in prostate cancer. AR is differentially expressed in the stroma and epithelium, with both paracrine and autocrine control throughout the prostate. Stromal-epithelial interactions within the prostate are commonly dependent on AR signaling and expression. Alterations in these pathways can promote tumorigenesis. AR is also expressed in normal and malignant mammary tissues. Emerging data indicate a role for AR in certain subtypes of breast cancer that has the potential to be exploited therapeutically. The aim of this review is to highlight the importance of these interactions in normal development and tumorigenesis, with a focus on the prostate and breast.


Subject(s)
Androgens/metabolism , Breast/metabolism , Epithelial Cells/metabolism , Prostate/metabolism , Stromal Cells/metabolism , Animals , Breast/growth & development , Humans , Male , Neoplasms/metabolism , Prostate/growth & development , Receptors, Androgen/metabolism , Signal Transduction
7.
Mol Endocrinol ; 27(7): 1048-64, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23744893

ABSTRACT

p21-Activated serine-threonine kinase (PAK1) is implicated in breast cancer. We have shown previously that PAK1 is tyrosyl phosphorylated by prolactin (PRL)-activated Janus tyrosine kinase (JAK2). Although a role for both PRL and PAK1 in breast cancer is widely acknowledged, the mechanism remains poorly understood. In the present study, PRL-activated PAK1 stimulates the invasion of TMX2-28 human breast cancer cells through Matrigel. Three-dimensional (3D) collagen IV stimulates the secretion of the matrix proteases, metalloproteinase (MMP)-1 and -3 that is further enhanced by the PRL-dependent tyrosyl phosphorylation of PAK1. 3D collagen IV also stimulates the expression and secretion of MMP-2, but in contrast to MMP-1 and -3, PRL/PAK1 signaling down-regulates MMP-2 expression and secretion. In contrast, MMP-9 expression and secretion are stimulated by 3D collagen I, not collagen IV, and are not affected by PRL but are down-regulated by PAK1. MMP-1 and -3 are required and MMP-2 contributes to PRL-dependent invasion. ERK1/2 signaling appears to be required for the enhanced expression and secretion of MMP-1 and -3 and enhanced PRL-dependent invasion. p38 MAPK and c-Jun N-terminal kinase 1/2 pathways participate in production of MMP-1 and -3 as well as in PRL/PAK1-dependent cell invasion. Together, these data illustrate the complex interaction between the substratum and PRL/PAK1 signaling in human breast cancer cells and suggest a pivotal role for PRL-dependent PAK1 tyrosyl phosphorylation in MMP secretion.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Collagen Type IV/pharmacology , Matrix Metalloproteinases/metabolism , Prolactin/pharmacology , p21-Activated Kinases/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Mutant Proteins/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Rats , Transcription, Genetic/drug effects
8.
Mol Endocrinol ; 27(3): 455-65, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23340249

ABSTRACT

The p21-activated serine-threonine kinase (PAK1) is activated by small GTPase-dependent and -independent mechanisms and regulates cell motility. Both PAK1 and the hormone prolactin (PRL) have been implicated in breast cancer by numerous studies. We have previously shown that the PRL-activated tyrosine kinase JAK2 (Janus tyrosine kinase 2) phosphorylates PAK1 in vivo and identified tyrosines (Tyr) 153, 201, and 285 in the PAK1 molecule as sites of JAK2 tyrosyl phosphorylation. Here, we have used human breast cancer T47D cells stably overexpressing PAK1 wild type or PAK1 Y3F mutant in which Tyr(s) 153, 201, and 285 were mutated to phenylalanines to demonstrate that phosphorylation of these three tyrosines are required for maximal PRL-dependent ruffling. In addition, phosphorylation of these three tyrosines is required for increased migration of T47D cells in response to PRL as assessed by two independent motility assays. Finally, we show that PAK1 phosphorylates serine (Ser) 2152 of the actin-binding protein filamin A to a greater extent when PAK1 is tyrosyl phosphorylated by JAK2. Down-regulation of PAK1 or filamin A abolishes the effect of PRL on cell migration. Thus, our data presented here bring some insight into the mechanism of PRL-stimulated motility of breast cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Movement/drug effects , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Phosphotyrosine/metabolism , Prolactin/pharmacology , p21-Activated Kinases/metabolism , Cell Line, Tumor , Clone Cells , Female , Filamins , Green Fluorescent Proteins/metabolism , Humans , Janus Kinase 2/metabolism , Models, Biological , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism
9.
Mol Endocrinol ; 25(9): 1565-78, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21719533

ABSTRACT

Prolactin (PRL) is critical for alveolar proliferation and differentiation in normal mammary development and is also implicated in breast cancer. PRL influences cell proliferation and growth by altering the expression of cyclin D1. Cyclin D1 expression is directly regulated by PRL through the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 5-mediated transcriptional activation of the cyclin D1 promoter. A p21-activated serine-threonine kinase (PAK)1 has also been implicated in the regulation of cyclin D1 gene expression. We have previously demonstrated that JAK2 directly phosphorylates PAK1 and extend these data here to demonstrate that PAK1 activates the cyclin D1 promoter in response to PRL. We show that mutation of PAK1 Tyr 153, 201, and 285 (sites of JAK2 phosphorylation; PAK1 Y3F) decreases both PAK1 nuclear translocation in response to PRL and PRL-induced cyclin D1 promoter activity by 55%. Mutation of the PAK1 nuclear localization signals decreases PRL-induced cyclin D1 promoter activity by 46%. A PAK1 Y3F mutant lacking functional nuclear localization signals decreases PRL-induced cyclin D1 activity by 68%, suggesting that there is another PAK1-dependent mechanism to activate the cyclin D1 promoter. We have found that adapter protein Nck sequesters PAK1 in the cytoplasm and that coexpression of both PAK1 and Nck inhibits the amplifying effect of PRL-induced PAK1 on cyclin D1 promoter activity (95% inhibition). This inhibition is partially abolished by disruption of PAK1-Nck binding. We propose two PAK1-dependent mechanisms to activate cyclin D1 promoter activity in response to PRL: via nuclear translocation of tyrosyl-phosphorylated PAK1 and via formation of a Nck-PAK1 complex that sequesters PAK1 in the cytoplasm.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclin D1/genetics , Oncogene Proteins/metabolism , Prolactin/pharmacology , Promoter Regions, Genetic , p21-Activated Kinases/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Humans , Models, Biological , Nuclear Localization Signals/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Transport/drug effects
10.
Mol Endocrinol ; 25(7): 1231-43, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21566085

ABSTRACT

Prolactin (PRL) regulates cytoskeletal rearrangement and cell motility. PRL-activated Janus tyrosine kinase 2 (JAK2) phosphorylates the p21-activated serine-threonine kinase (PAK)1 and the Src homology 2 (SH2) domain-containing adapter protein SH2B1ß. SH2B1ß is an actin-binding protein that cross-links actin filaments, whereas PAK1 regulates the actin cytoskeleton by different mechanisms, including direct phosphorylation of the actin-binding protein filamin A (FLNa). Here, we have used a FLNa-deficient human melanoma cell line (M2) and its derivative line (A7) that stably expresses FLNa to demonstrate that SH2B1ß and FLNa are required for maximal PRL-dependent cell ruffling. We have found that in addition to two actin-binding domains, SH2B1ß has a FLNa-binding domain (amino acids 200-260) that binds directly to repeats 17-23 of FLNa. The SH2B1ß-FLNa interaction participates in PRL-dependent actin rearrangement. We also show that phosphorylation of the three tyrosines of PAK1 by JAK2, as well as the presence of FLNa, play a role in PRL-dependent cell ruffling. Finally, we show that the actin- and FLNa-binding-deficient mutant of SH2B1ß (SH2B1ß 3Δ) abolished PRL-dependent ruffling and PRL-dependent cell migration when expressed along with PAK1 Y3F (JAK2 tyrosyl-phosphorylation-deficient mutant). Together, these data provide insight into a novel mechanism of PRL-stimulated regulation of the actin cytoskeleton and cell motility via JAK2 signaling through FLNa, PAK1, and SH2B1ß. We propose a model for PRL-dependent regulation of the actin cytoskeleton that integrates our findings with previous studies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Contractile Proteins/metabolism , Cytoskeleton/metabolism , Microfilament Proteins/metabolism , Prolactin/pharmacology , Actins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Motifs , Binding Sites , Cell Line, Tumor , Cell Surface Extensions/metabolism , Filamins , Humans , Janus Kinase 2/metabolism , Phosphorylation , Prolactin/physiology , Protein Binding , Protein Isoforms/metabolism , Protein Transport , p21-Activated Kinases/metabolism
11.
Mol Endocrinol ; 23(7): 1065-76, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19342444

ABSTRACT

The Src homology 2 (SH2) domain-containing adapter protein SH2B1beta plays a role in severe obesity, leptin and insulin resistance, and infertility. SH2B1beta was initially identified as a Janus tyrosine kinase 2 (JAK2) substrate, and it has been implicated in cell motility and regulation of the actin rearrangement in response to GH and platelet-derived growth factor. SH2B1beta is also required for maximal actin-based motility of Listeria. Here we have used a low-speed pelleting assay and electron microscopy to demonstrate that SH2B1beta has two actin-binding sites and that it cross-links actin filaments in vitro. Wild-type SH2B1beta localized to cell ruffles and along filopodia, but deletion of amino acids 150-200 (the first actin-binding site) led to mislocalization of the protein to filopodia tip complexes where it colocalized with vasodilator-stimulated phosphoprotein (VASP). Based on studies performed in VASP-deficient MVD7(-/-) cells, with or without green fluorescent protein-VASP reconstitution, we concluded that the proper intracellular localization of native SH2B1beta required the presence of the first SH2B1beta actin-binding site and VASP. Finally, we found that both SH2B1beta actin-binding domains were required for maximal GH- and prolactin-induced cell ruffling. Together, these results suggest that SH2B1beta functions as an adapter protein that cross-links actin filaments, leading to modulation of cellular responses in response to JAK2 activation.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/physiology , Cytoskeleton/metabolism , Protein Multimerization/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Movement/physiology , Cells, Cultured , Humans , Janus Kinase 2/metabolism , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Microvilli/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutant Proteins/physiology , Phosphoproteins/metabolism , Phosphoproteins/physiology , Protein Binding/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Pseudopodia/metabolism
12.
J Biol Chem ; 282(42): 30985-96, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17726028

ABSTRACT

The serine-threonine kinase PAK1 is activated by small GTPase-dependent and -independent mechanisms and promotes cell survival. However, the role of tyrosyl phosphorylation in the regulation of PAK1 function is poorly understood. In this study, we have shown that the prolactin-activated tyrosine kinase JAK2 phosphorylates PAK1 in vivo. Wild type, but not kinase-dead, JAK2 directly phosphorylates PAK1 in cells and in an in vitro kinase assay. PAK1 tyrosines 153, 201, and 285 were identified as sites of JAK2 tyrosyl phosphorylation by mass spectrometry and two-dimensional peptide mapping. Mutation of PAK1 tyrosines 153, 201, and 285 to phenylalanines individually or in combination implicated these PAK1 tyrosines in the regulation of PAK1 kinase activity. Tyrosyl phosphorylation by JAK2 significantly increases PAK1 kinase activity, whereas similar phosphorylation of the PAK1 Y153F,Y201F,Y285F mutant has no effect on PAK1 activity. Tyrosyl phosphorylation of wild type PAK1 decreases apoptosis induced by serum deprivation and staurosporine treatment and increases cell motility. In contrast, these parameters are unaltered in the PAK1 Y153F,Y201F,Y285F mutant. Our findings indicate that JAK2 phosphorylates PAK1 at these specific tyrosines and that this phosphorylation plays an important role in cell survival and motility.


Subject(s)
Apoptosis/physiology , Cell Movement/physiology , Janus Kinase 2/metabolism , Protein Processing, Post-Translational/physiology , p21-Activated Kinases/metabolism , Amino Acid Substitution , Animals , Apoptosis/drug effects , COS Cells , Cell Movement/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Chlorocebus aethiops , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , Janus Kinase 2/genetics , Mutation, Missense , Peptide Mapping , Phosphorylation/drug effects , Prolactin/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Staurosporine/pharmacology , p21-Activated Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...