Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mol Pharm ; 21(9): 4648-4663, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39152916

ABSTRACT

Occult nodal spread and metastatic disease require longstanding imaging and biochemical assessments for thyroid cancer, a disease that has a propensity for diffuse, small-volume disease. We have developed a 64Cu-labeled platelet-derived growth factor receptor α (PDGFRA) antibody for immuno-PET of PDGFRA in metastatic papillary thyroid cancer (PTC). The present work describes the discovery of small cyclic PDGFRA-targeting peptides, their binding features, and radiolabeling with positron emitter gallium-68 (68Ga) for in vitro and in vivo characterization in thyroid cancer models. Phage-display technology with two separate libraries and seven different cell lines was used through three rounds of biopanning as well as flow cytometry and comparative analysis with recombinant protein to select specific peptide sequences. Phenotypic binding analysis was completed by using phosphorylation and cell migration assays. In vitro protein binding was analyzed with thermophoresis and flow cytometry using the fluorescent-labeled PDGFRA peptide. Peptide candidates were modified with the NOTA chelator for radiolabeling with 68Ga. In vitro cell uptake was studied in various thyroid cancer cell lines. In vivo studies of 68Ga-labeled peptides included metabolic stability and PET imaging. From the original library (1013 compounds), five different peptide groups were identified based on biopanning experiments with and without the α subunit of PDGFR, leading to ∼50 peptides. Subsequent phenotypic screening revealed two core peptide sequences (CP16 and CP18) that demonstrated significant changes in the level of PDGFRA phosphorylation and cell migration. Alanine scan sublibraries were created from these two lead peptide sequences, and peptides were radiolabeled using 68Ga-GaCl3 at pH 4.5, resulting in RCP > 95% within 34-40 min, including SPE purification. Cyclic peptide CP18.5 showed the strongest effects on cell migration, flow cytometry, and binding by visual interference color assay. 68Ga-labeled PDGFRA-targeting peptides showed elevated cell and tumor uptake in models of thyroid cancer, with 68Ga-NOTA-CP18.5 being the lead candidate. However, metabolic stability in vivo was compromised for 68Ga-NOTA-CP18.5 vs 68Ga-NOTA-CP18 but without impacting tumor uptake or clearance profiles. First-generation radiolabeled cyclic peptides have been developed as novel radiotracers, particularly 68Ga-NOTA-CP18.5, for the molecular imaging of PDGFRA in thyroid cancer.


Subject(s)
Gallium Radioisotopes , Molecular Imaging , Peptides, Cyclic , Receptor, Platelet-Derived Growth Factor alpha , Humans , Animals , Cell Line, Tumor , Mice , Peptides, Cyclic/chemistry , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Gallium Radioisotopes/chemistry , Molecular Imaging/methods , Positron-Emission Tomography/methods , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Thyroid Cancer, Papillary/diagnostic imaging , Thyroid Cancer, Papillary/metabolism , Cell Movement , Copper Radioisotopes/chemistry , Mice, Nude , Female
2.
Endocrine ; 72(2): 452-461, 2021 05.
Article in English | MEDLINE | ID: mdl-32914379

ABSTRACT

PURPOSE: Platelet derived growth receptor alpha (PDGFRA) promotes the epithelial-mesenchymal transition (EMT) in thyroid follicular cells and is linked to lymphatic metastases in papillary thyroid cancer (PTC). We probed the regulatory network of genes linked to PDGFRA and EMT, comparing matched patient primary tumor and metastatic specimens, as well as engineered cell lines and ex vivo primary cultures with and without PDGFRA. METHODS: Freshly isolated thyroid tumors with or without metastases, with matching neighboring benign or normal tissue, was isolated for comparative transcriptional analysis using a TaqMan Low Density array (TLDA) assay with genes representing important markers of EMT, cellular adhesion, apoptosis, differentiation, senescence, and signal transduction pathways in thyroid cancer. Transfected primary cultures and immortalized cell lines were also analyzed with respect to PDGFRA expression and cell phenotype. RESULTS: We reveal the consistent upregulation of serine protease DPP4 and structural protein SPP1 with the progression of PTC to metastatic disease, as well as with PDGFRA expression. Conversely, epithelial integrity gene TFF3 and transcription factor SOX10 were strongly down-regulated. This gene network also includes important mediators of EMT including DSG1, MMP3, MMP9, and BECN. We observed similar genomic changes in ex vivo normal thyroid cells transfected with PDGFRA that also exhibited a partially dedifferentiated phenotype. In particular, we observed lamellopodia with induction of PDGFRA and illustrate that DPP4 and SPP1 were upregulated in this process, with decreased TFF3 and SOX10 as seen in tissue specimens. PDGFRA did decrease nuclear protein levels of differentiation factor TTF1, but not the transcription of TTF1 and PAX8. CONCLUSIONS: We demonstrate that PDGFRA activates EMT pathways and decreases expression of genes favoring epithelial integrity, pushing follicular cells toward a dedifferentiated phenotype. SPP1 and DPP4, previously linked with adverse outcomes in thyroid cancer, appear to be regulated by PDGFRA. PDGFRA expression promotes metastatic disease through multiple EMT levers that favor formation of an invasive phenotype and increased metalloproteinase expression.


Subject(s)
Epithelial-Mesenchymal Transition , Thyroid Neoplasms , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Transcriptome
3.
Endocr Relat Cancer ; 27(9): 495-507, 2020 09.
Article in English | MEDLINE | ID: mdl-32590338

ABSTRACT

Targeted therapy is increasingly used to manage metastatic papillary thyroid cancer. The focus of the present study was to examine glucose metabolism and tumor responses for thyroid cancer xenografts expressing the glycolytic pathway modulators platelet-derived growth factor receptor (PDGFR) and BRAFV600E. Radiolabelled glucose derivative [18F]FDG was used to analyze the effects of PDGFR blockade with imatinib, BRAF blockade with vemurafenib, as well as combined PDGFR and BRAF blockade in vitro and in vivo with PET. Dynamic PET data was correlated with immunohistochemistry staining and kinetic analysis for facilitative glucose transporter 1 (GLUT1) and hexokinase-II (HK2). Vemurafenib decreased [18F]FDG uptake in BCPAP cells in vitro; however, it was increased by ~70% with imatinib application to BCPAP cells. This metabolic response to tyrosine kinase inhibition required BRAFV600E as it was not seen in cell lines lacking mutated BRAF (TPC1). In xenografts, imatinib therapy in BCPAP thyroid tumour-bearing mice significantly increased [18F]FDG uptake and retention (>30%) in BCPAP tumours with PDGFRß or both (α+ß) isoforms. Kinetic analysis revealed that the increased glucose uptake is a consequence of increased phosphorylation and intracellular trapping of [18F]FDG confirmed by an increase in HK2 protein expression and activity, but not GLUT1 activity. BRAF inhibition alone, or combined PDGFR and BRAF inhibition, reduced (~60%) [18F]FDG uptake in both types of BCPAP (ß or α+ß) tumours. In terms of tumour growth, combination therapy with imatinib and vemurafenib led to a near abolition of the tumors (~90% reduction), but single therapy for BCPAP with PDGFRα expression was much less effective. In summary, imatinib led to a paradoxical increase of [18F]FDG uptake in xenografts that was reversed through BRAFV600E inhibition. The present data show that metabolic reprogramming in thyroid cancer occurs as a consequence of BRAF-mediated upregulation of HK2 expression that may permit tumour growth with isolated blockade of upstream tyrosine kinase receptors.


Subject(s)
Positron Emission Tomography Computed Tomography/methods , Protein Kinase Inhibitors/therapeutic use , Thyroid Cancer, Papillary/drug therapy , Animals , Fluorodeoxyglucose F18/therapeutic use , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Thyroid Cancer, Papillary/pathology
4.
Hum Pathol ; 91: 36-42, 2019 09.
Article in English | MEDLINE | ID: mdl-31229486

ABSTRACT

Function of the thyroid follicular cell depends on nuclear expression of thyroid transcription factor 1 (TTF1). Regulation of this key protein regulating iodide transport is not well known, but its loss is linked to the most lethal of thyroid malignancies. We examined TTF1 nuclear expression in the context of adverse pathological features, disease recurrence, and BRAF status in papillary thyroid carcinomas with (n = 182) and without (n = 303) nodal metastases. Overall nuclear expression level of TTF1 was strong and diffuse in approximately 73%, whereas 27% exhibited lower levels or a paucity of nuclear staining. In the same cohort, approximately 59% exhibited the BRAF mutation. On univariate analysis, low levels of TTF1 nuclear expression was linked to vascular invasion, extrathyroidal extension, and nodal metastases. Multivariate analysis indicated that low levels of TTF1 were most strongly linked to nodal metastases and vascular invasion. Interestingly, TTF1 levels were not linked to the BRAF mutation. TTF1 staining alone predicted disease recurrence, but when combined with BRAF status, the 2 markers exhibited a more marked influence. Patients lacking the BRAF mutation and exhibiting normal levels of TTF1 exhibited very low levels of disease recurrence (11% at 10 years). Conversely, patient tumors with low levels of TTF1 and the BRAF mutation recurred in 31% of cases in the same time frame. The mixed expression of BRAF under varying levels of differentiation may explain, in part, the contradictory studies regarding the impact of BRAF mutations on patient prognosis and also indicates a complex genomic signature for dedifferentiated thyroid cancer.


Subject(s)
Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , Thyroid Nuclear Factor 1/metabolism , Adult , Cohort Studies , DNA-Binding Proteins , Disease-Free Survival , Female , Humans , Male , Middle Aged , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Proto-Oncogene Proteins B-raf/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Transcription Factors
5.
Hum Pathol ; 75: 146-153, 2018 05.
Article in English | MEDLINE | ID: mdl-29408504

ABSTRACT

Metastatic dissemination of papillary thyroid cancer has been reported to be strongly associated with expression of platelet-derived growth factor (PDGFR) α and altered TTF1 function. However, the status of PDGF ligands in papillary thyroid cancer and the potential role of these ligands in metastatic disease are obscure. We assessed the prevalence of PDGF ligands in benign and malignant thyroid tumors to determine if ligand upregulation is associated with α-isoform (PDGF-AA or PDGF-BB) or the ß-isoform (PDGF-BB or PDGF-DD) of PDGFR in individual tumors. The immunohistochemical expression of PDGFRα, PDGF-AA, PDGF-BB, and PDGF-DD was surveyed in follicular adenomas (n=55), papillary thyroid carcinomas (103 with and 59 without nodal metastases), and lymph node metastasis (n=12). There is an augmented tendency for PDGF-AA expression in node-positive papillary thyroid cancer metastases (P<.0001). Although PDGF-BB and -DD were commonly identified, there was no relationship between the presence of these cytokines and malignant disease or metastases. Logistic regression demonstrated that PDGF-AA expression was significantly associated with the presence of PDGFRα (odds ratio=4.6, P=.004) and recurrent disease. When either PDGFRα or PDGF-AA was used to predict the presence of metastases, the sensitivity achieved was 86% and 88%, respectively, whereas specificities were lower at 71% and 61%, respectively. The augmented coexpression of PDGF-AA and PDGFRα in metastatic papillary thyroid cancers suggests that an autocrine signaling loop may contribute to nodal infiltration. Combined testing for the expression of PDGF-AA and PDGFRα may identify those patients with papillary thyroid cancer at risk of metastatic disease and resistance to therapy.


Subject(s)
Autocrine Communication/physiology , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/analysis , Female , Humans , Lymphatic Metastasis/pathology , Male , Middle Aged , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/metabolism
6.
Nucl Med Biol ; 58: 51-58, 2018 03.
Article in English | MEDLINE | ID: mdl-29367096

ABSTRACT

INTRODUCTION: Receptor tyrosine kinase (RTK) platelet-derived growth factor receptor-alpha (PDGFRα) was recently identified as a molecular switch for dedifferentiation in thyroid cancer that predicts resistance to therapy as well as recurrence of disease in papillary thyroid cancer. Here we describe the radiolabeling and functional characterization of an imaging probe based on a PDGFRα-specific monoclonal antibody (mAb) for immuno-PET imaging of PDGFRα in papillary thyroid cancer. METHODS: Antibody D13C6 (Cell Signaling) was decorated with chelator NOTA using bioconjugation reaction with 2-(p-NCS-Bz)-NOTA. Radiolabeling was carried out using 40 µg of antibody-NOTA conjugate with 143-223 MBq of [64Cu]CuCl2 in 0.25 M NaOAc (pH 5.5) at 30 °C for 1 h. The reaction mixture was purified with size-exclusion chromatography (PD-10 column). PDGFRα and mock transfected B-CPAP thyroid cancer cells lines for validation of 64Cu-labeled immuno-conjugates were generated using LVX-Tet-On technology. PET imaging was performed in NSG mice bearing bilaterally-induced PDGFRα (+/-) B-CPAP tumors. RESULTS: Bioconjugation of NOTA chelator to monoclonal antibody D13C6 resulted in 2.8 ±â€¯1.3 chelator molecules per antibody as determined by radiometric titration with 64Cu. [64Cu]Cu-NOTA-D13C6 was isolated in high radiochemical purity (>98%) and good radiochemical yields (19-61%). The specific activity was 0.9-5.1 MBq/µg. Cellular uptake studies revealed a specific radiotracer uptake in PDGFRα expressing cells compared to control cells. PET imaging resulted in SUVmean values of ~5.5 for PDGFRα (+) and ~2 for PDGFRα (-) tumors, after 48 h p.i.. After 1 h, radiotracer uptake was also observed in the bone marrow (SUVmean ~5) and spleen (SUVmean ~8.5). CONCLUSION: Radiolabeled antibody [64Cu]Cu-NOTA-D13C6 represents a novel and promising radiotracer for immuno-PET imaging of PDGFRα in metastatic papillary thyroid cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: The presented work has the potential to allow physicians to identify papillary thyroid cancer patients at risk of metastases by using the novel immuno-PET imaging assay based on PDGFRα-targeting antibody [64Cu]Cu-NOTA-D13C6.


Subject(s)
Positron-Emission Tomography/methods , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Thyroid Cancer, Papillary/diagnostic imaging , Animals , Autoradiography , Biological Transport , Cell Line, Tumor , Copper Radioisotopes , Heterocyclic Compounds/chemistry , Heterocyclic Compounds, 1-Ring , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Mice , Thyroid Cancer, Papillary/immunology , Thyroid Cancer, Papillary/metabolism
7.
Oncotarget ; 7(50): 83684-83700, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27845909

ABSTRACT

Recently platelet derived growth factor receptor-alpha (PDGFRα) was recognized as a potential target to treat aggressive papillary thyroid cancer given its strong association with lymph node metastases. However, it is unclear how PDGFRα potentiates metastases and if it works through the canonical MAPK pathway traditionally linked to PTC oncogenesis. We explored the phenotypic changes driven by PDGFRα activation in human papillary thyroid cancer (PTC) cells and the downstream signalling cascades through which they are effected. We demonstrate that PDGFRα drives an impressive phenotypic change in PTC cell lines as documented by significant cytoskeletal rearrangement, increased migratory potential, and the formation of invadopodia. Cells lacking PDGFRα formed compact and dense spheroids, whereas cells expressing active PDGFRα exhibited invadopodia in three-dimensional culture. To achieve this, active PDGFRα provoked downstream activation of the MAPK/Erk, PI3K/Akt and STAT3 pathways. We further confirmed the role of PDGFRα as a transformative agent promoting the epithelial to mesenchymal transition of PTC cells, through the augmentation of Snail and Slug expression. Crenolanib, a small molecule inhibitor of PDGFRα, suppressed the levels of Snail and Slug and almost completely reversed all the phenotypic changes. We demonstrate that PDGFRα activation is an essential component that drives aggressiveness in PTC cells, and that the signaling pathways are complex, involving not only the MAPK/Erk but also the PI3K/Akt and STAT3 pathways. This argues for upstream targeting of the PDGFRα given the redundancy of oncogenic pathways in PTC, especially in patients whose tumors over-express this tyrosine kinase receptor.


Subject(s)
Carcinoma, Papillary/metabolism , Epithelial-Mesenchymal Transition , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Thyroid Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/genetics , Carcinoma, Papillary/secondary , Cell Line, Tumor , Cell Movement , Cytoskeleton/metabolism , Cytoskeleton/pathology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lymphatic Metastasis , Phenotype , Phosphatidylinositol 3-Kinase/metabolism , Piperidines/pharmacology , Podosomes/metabolism , Podosomes/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Snail Family Transcription Factors/metabolism , Thyroid Cancer, Papillary , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Time Factors
8.
EBioMedicine ; 12: 86-97, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27682510

ABSTRACT

Dedifferentiation of follicular cells is a central event in resistance to radioactive iodine and patient mortality in papillary thyroid carcinoma (PTC). We reveal that platelet derived growth factor receptor alpha (PDGFRα) specifically drives dedifferentiation in PTC by disrupting the transcriptional activity of thyroid transcription factor-1 (TTF1). PDGFRα activation dephosphorylates TTF1 consequently shifting the localization of this transcription factor from the nucleus to the cytoplasm. TTF1 is required for follicular cell development and disrupting its function abrogates thyroglobulin production and sodium iodide transport. PDGFRα also promotes a more invasive and migratory cell phenotype with a dramatic increase in xenograft tumor formation. In patient tumors we confirm that nuclear TTF1 expression is inversely proportional to PDGFRα levels. Patients exhibiting PDGFRα at time of diagnosis are three times more likely to exhibit nodal metastases and are 18 times more likely to recur within 5years than those patients lacking PDGFRα expression. Moreover, high levels of PDGFRα and low levels of nuclear TTF1 predict resistance to radioactive iodine therapy. We demonstrate in SCID xenografts that focused PDGFRα blockade restores iodide transport and decreases tumor burden by >50%. Focused PDGFRα inhibitors, combined with radioactive iodine, represent an additional avenue for treating patients with aggressive variants of PTC.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , Drug Resistance, Neoplasm/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Animals , Biological Transport , Carcinoma/drug therapy , Carcinoma/mortality , Carcinoma, Papillary , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Models, Biological , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Recurrence, Local , Phenotype , Prognosis , Protein Transport , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sodium Iodide/metabolism , Thyroglobulin/biosynthesis , Thyroid Cancer, Papillary , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/mortality , Transcription Factors , Xenograft Model Antitumor Assays
9.
Endocr Relat Cancer ; 22(4): 593-607, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26037280

ABSTRACT

Autotaxin is a secreted enzyme that converts extracellular lysophosphatidylcholine to lysophosphatidate (LPA). In cancers, LPA increases tumour growth, metastasis and chemoresistance by activating six G-protein coupled receptors. We examined >200 human thyroid biopsies. Autotaxin expression in metastatic deposits and primary carcinomas was four- to tenfold higher than in benign neoplasms or normal thyroid tissue. Autotaxin immunohistochemical staining was also increased in benign neoplasms with leukocytic infiltrations. Malignant tumours were distinguished from benign tumours by high tumour autotaxin, LPA levels and inflammatory mediators including IL1ß, IL6, IL8, GMCSF, TNFα, CCL2, CXCL10 and platelet-derived growth factor (PDGF)-AA. We determined the mechanistic explanation for these results and revealed a vicious regulatory cycle in which LPA increased the secretion of 16 inflammatory modulators in papillary thyroid cancer cultures. Conversely, treating cancer cells with ten inflammatory cytokines and chemokines or PDGF-AA and PDGF-BB increased autotaxin secretion. We confirmed that this autotaxin/inflammatory cycle occurs in two SCID mouse models of papillary thyroid cancer by blocking LPA signalling using the autotaxin inhibitor ONO-8430506. This decreased the levels of 16 inflammatory mediators in the tumours and was accompanied by a 50-60% decrease in tumour volume. This resulted from a decreased mitotic index for the cancer cells and decreased levels of vascular endothelial growth factor and angiogenesis in the tumours. Our results demonstrate that the autotaxin/inflammatory cycle is a focal point for driving malignant thyroid tumour progression and possibly treatment resistance. Inhibiting autotaxin activity provides an effective and novel strategy for decreasing the inflammatory phenotype in thyroid carcinomas, which should complement other treatment modalities.


Subject(s)
Inflammation Mediators/metabolism , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Thyroid Neoplasms/metabolism , Adult , Aged , Animals , Female , Humans , Male , Mice , Mice, SCID , Middle Aged , Thyroid Gland/metabolism , Thyroid Neoplasms/pathology
10.
PLoS One ; 8(8): e72331, 2013.
Article in English | MEDLINE | ID: mdl-23991094

ABSTRACT

Ingenol-3-angelate (I3A) is a non-tumor promoting phorbol ester-like compound identified in the sap of Euphoria peplus. Similar to tumor promoting phorbol esters, I3A is a diacylglycerol (DAG) analogue that binds with high affinity to the C1 domains of PKCs, recruits PKCs to cellular membranes and promotes enzyme activation. Numerous anti-cancer activities have been attributed to I3A and ascribed to I3A's effects on PKCs. We show here that I3A also binds to and activates members of the RasGRP family of Ras activators leading to robust elevation of Ras-GTP and engagement of the Raf-Mek-Erk kinase cascade. In response to I3A, recombinant proteins consisting of GFP fused separately to full-length RasGRP1 and RasGRP3 were rapidly recruited to cell membranes, consistent with direct binding of the compound to RasGRP's C1 domain. In the case of RasGRP3, IA3 treatment led to positive regulatory phosphorylation on T133 and activation of the candidate regulatory kinase PKCδ. I3A treatment of select B non-Hodgkin's lymphoma cell lines resulted in quantitative and qualitative changes in Bcl-2 family member proteins and induction of apoptosis, as previously demonstrated with the DAG analogue bryostatin 1 and its synthetic analogue pico. Our results offer further insights into the anticancer properties of I3A, support the idea that RasGRPs represent potential cancer therapeutic targets along with PKC, and expand the known range of ligands for RasGRP regulation.


Subject(s)
Antineoplastic Agents/pharmacology , Diterpenes/pharmacology , ras Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Protein Kinases/metabolism , Rats , ras Proteins/metabolism
11.
Methods Mol Biol ; 1005: 129-41, 2013.
Article in English | MEDLINE | ID: mdl-23606254

ABSTRACT

We describe various complementary techniques to achieve multidimensional mitochondrial proteome fractionation and analysis. Previously described methods for 2D-DIGE/mass spectrometry and 1D-SDS-PAGE/Western techniques and protein complex analysis by BN-PAGE/Western and sucrose gradient ultracentrifugation/SDS-PAGE/mass spectrometry are optimized to characterize the brain mitochondrial proteome. This approach allows for a comprehensive identification of mitochondrial proteomic differences between health and disease conditions.


Subject(s)
Brain/metabolism , Mitochondria/chemistry , Peptide Mapping/methods , Proteome/chemistry , Animals , Brain Chemistry , Centrifugation, Density Gradient , Chemical Fractionation , Electrophoresis, Polyacrylamide Gel , Gene Expression , Humans , Isoelectric Focusing , Mitochondria/genetics , Mitochondria/metabolism , Proteolysis , Proteome/genetics , Proteome/metabolism , Rats , Tandem Mass Spectrometry
12.
Exp Hematol ; 40(8): 646-56.e2, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22465296

ABSTRACT

The anti-cancer effects of bryostatin-1, a potent diacylglycerol analogue, have traditionally been attributed to its action on protein kinase C. However, we previously documented apoptosis in a B non-Hodgkin lymphoma cell line involving diacylglycerol analogue stimulation of Ras guanyl-releasing protein, a Ras activator, and Bim, a proapoptotic Bcl-2 family protein. To further explore the role of Bim, we examined several Bim-deficient B non-Hodgkin lymphoma cells for their responses to pico, a synthetic bryostatin-1-like compound. The Bim(-) mantle cell lymphoma cell lines Jeko-1, Mino, Sp53, UPN1, and Z138 and the Bim(+) cell line Rec-1, as well as the Burkitt lymphoma cells lines BL2 (Bim(-)) and Daudi (Bim(+)), were examined for their response to pico using assays for proliferation and apoptosis as well as biochemical methods for Ras guanyl-releasing proteins and Bcl-2 family members. With the exception of UPN1, mantle cell lymphoma cell lines underwent pico-induced apoptosis, as did BL2. In some cases, hallmarks of apoptosis were substantially diminished in the presence of mitogen-activated protein kinase kinase inhibitors. Pico treatment generally led to increased expression of proapoptotic Bik, although the absolute levels of Bik varied considerably between cell lines. A pico-resistant variant of Z138 exhibited decreased Bik induction compared to parental Z138 cells. Pico also generally decreased expression of anti-apoptotic Bcl-XL and Mcl1. Although, these changes in Bcl-2 family members seem unlikely to fully account for the differential behavior of the cell lines, our demonstration of a potent apoptotic process in most cell lines derived from mantle cell lymphoma encourages a re-examination of diacylglycerol analogues in the treatment of this subset of B non-Hodgkin lymphoma cases.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/physiology , Apoptosis/drug effects , Bryostatins/pharmacology , Lymphoma, Mantle-Cell/drug therapy , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Bcl-2-Like Protein 11 , Cell Line, Tumor , DNA-Binding Proteins/analysis , DNA-Binding Proteins/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanine Nucleotide Exchange Factors/analysis , Guanine Nucleotide Exchange Factors/physiology , Humans , Lymphoma, Mantle-Cell/pathology , Proto-Oncogene Proteins c-bcl-2/analysis , bcl-X Protein/analysis
13.
Circulation ; 119(18): 2480-9, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19398663

ABSTRACT

BACKGROUND: Excessive stimulation of Gq protein-coupled receptors by cognate vasoconstrictor agonists induces a variety of cardiovascular processes, including hypertension and hypertrophy. Here, we report that matrix metalloproteinase-7 (MMP-7) and a disintegrin and metalloproteinase-12 (ADAM-12) form a novel signaling axis in these processes. METHODS AND RESULTS: In functional studies, we targeted MMP-7 in rodent models of acute, long-term, and spontaneous hypertension by 3 complementary approaches: (1) Pharmacological inhibition of activity, (2) expression knockdown (by antisense oligodeoxynucleotides and RNA interference), and (3) gene knockout. We observed that induction of acute hypertension by vasoconstrictors (ie, catecholamines, angiotensin II, and the nitric oxide synthase inhibitor N(G)-nitro-l-arginine methyl ester) required the posttranscriptional activation of vascular MMP-7. In spontaneously hypertensive rats, knockdown of MMP-7 (by RNA interference) resulted in attenuation of hypertension and stopped development of cardiac hypertrophy. Quantitative reverse-transcription polymerase chain reaction studies in mouse models of MMP-7 knockdown (by RNA interference) and gene knockout revealed that MMP-7 controlled the transcription of ADAM-12, the major metalloproteinase implicated in cardiac hypertrophy. In mice with angiotensin II-induced hypertension and cardiac hypertrophy, myocardial ADAM-12 and downstream hypertrophy marker genes were overexpressed. Knockdown of MMP-7 attenuated hypertension, inhibited ADAM-12 overexpression, and prevented cardiac hypertrophy. CONCLUSIONS: Agonist signaling of both hypertension and hypertrophy depends on posttranscriptional and transcriptional mechanisms that involve MMP-7, which is transcriptionally connected with ADAM-12. Approaches targeting this novel MMP-7/ADAM-12 signaling axis could have generic therapeutic potential in hypertensive disorders caused by multiple or unknown agonists.


Subject(s)
ADAM Proteins/metabolism , Cardiomegaly/metabolism , Hypertension/metabolism , Matrix Metalloproteinase 7/metabolism , Signal Transduction/physiology , ADAM Proteins/genetics , ADAM12 Protein , Acute Disease , Adrenergic alpha-Agonists/pharmacology , Animals , Cardiomegaly/physiopathology , Disease Models, Animal , Hypertension/chemically induced , Hypertension/physiopathology , Matrix Metalloproteinase 7/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Norepinephrine/pharmacology , Phenylephrine/pharmacology , RNA Interference , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley
14.
Exp Hematol ; 37(1): 122-134, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19100522

ABSTRACT

OBJECTIVE: Bryostatin-1 and related diacylglycerol (DAG) analogues activate RasGRPs in lymphocytes, thereby activating Ras and mimicking some aspects of immune receptor signaling. To define the role of RasGRPs in lymphocyte apoptosis and to identify potential therapeutic uses for DAG analogues in lymphocyte disorders, we characterized the response of lymphoma-derived cell lines to DAG analogues. MATERIALS AND METHODS: Human lymphoma-derived B cell lines and mouse primary B cells were treated with bryostatin-1 or its synthetic analogue "pico." Ras signaling partners and Bcl-2 family members were studied with biochemical assays. Cellular responses were monitored using growth and apoptosis assays. RESULTS: Stimulation of B cells with DAG analogues results in activation of protein kinase C/RasGRP-Ras-Raf-Mek-Erk signaling and phosphorylation of the proapoptotic BH3-only protein Bim. In vitro, Bim is phosphorylated by Erk on sites previously associated with increased apoptotic activity. In Toledo B cells derived from a non-Hodgkin's lymphoma (B-NHL), DAG analogue stimulation leads to extensive apoptosis. Apoptosis can be suppressed by either downregulation of Bim or overexpression of Bcl-2. It is associated with the formation of Bak-Bax complexes and increased mitochondrial membrane permeability. Toledo B-NHL cell apoptosis shows a striking dependence on sustained signaling. CONCLUSION: In B cells, Erk activation leads directly to phosphorylation of Bim on sites associated with activation of Bim. In Toledo B-NHL cells, the dependence of apoptosis on sustained signaling suggests that Bcl-2 family members could interpret signal duration, an important determinant of B cell receptor-mediated negative selection. Certain cases of B-NHL might respond to DAG analogue treatment by the mechanism outlined here.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , B-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , MAP Kinase Signaling System/physiology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bcl-2-Like Protein 11 , Bryostatins/pharmacology , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Humans , MAP Kinase Signaling System/drug effects , Mice , Mitochondrial Membranes/metabolism , Phosphorylation/drug effects , Receptors, Antigen, B-Cell/metabolism , bcl-2-Associated X Protein/metabolism
15.
Biophys J ; 94(10): 3748-59, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18234819

ABSTRACT

We present a general-purpose model for biomolecular simulations at the molecular level that incorporates stochasticity, spatial dependence, and volume exclusion, using diffusing and reacting particles with physical dimensions. To validate the model, we first established the formal relationship between the microscopic model parameters (timestep, move length, and reaction probabilities) and the macroscopic coefficients for diffusion and reaction rate. We then compared simulation results with Smoluchowski theory for diffusion-limited irreversible reactions and the best available approximation for diffusion-influenced reversible reactions. To simulate the volumetric effects of a crowded intracellular environment, we created a virtual cytoplasm composed of a heterogeneous population of particles diffusing at rates appropriate to their size. The particle-size distribution was estimated from the relative abundance, mass, and stoichiometries of protein complexes using an experimentally derived proteome catalog from Escherichia coli K12. Simulated diffusion constants exhibited anomalous behavior as a function of time and crowding. Although significant, the volumetric impact of crowding on diffusion cannot fully account for retarded protein mobility in vivo, suggesting that other biophysical factors are at play. The simulated effect of crowding on barnase-barstar dimerization, an experimentally characterized example of a bimolecular association reaction, reveals a biphasic time course, indicating that crowding exerts different effects over different timescales. These observations illustrate that quantitative realism in biosimulation will depend to some extent on mesoscale phenomena that are not currently well understood.


Subject(s)
Biopolymers/chemistry , Colloids/chemistry , Cytoplasm/chemistry , Models, Biological , Models, Chemical , Models, Molecular , Computer Simulation , Diffusion , Kinetics
16.
Hypertension ; 51(2): 412-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18172056

ABSTRACT

The central nervous system plays a critical role in the normal control of arterial blood pressure and in its elevation in virtually all forms of hypertension. Mitochondrial dysfunction has been increasingly associated with the development of hypertension. Therefore, we examined whether mitochondrial dysfunction occurs in the brain in hypertension and characterized it at the molecular scale. Mitochondria from whole brain and brain stem from 12-week-old spontaneously hypertensive rats with elevated blood pressure (190+/-5 mm Hg) were compared against those from age-matched normotensive (134+/-7 mm Hg) Wistar Kyoto rats (n=4 in each group). Global differential analysis using 2D electrophoresis followed by tandem mass spectrometry-based protein identification suggested a downregulation of enzymes involved in cellular energetics in hypertension. Targeted differential analysis of mitochondrial respiratory complexes using the classical blue-native SDS-PAGE/Western method and a complementary combination of sucrose-gradient ultracentrifugation/tandem mass spectrometry revealed previously unknown assembly defects in complexes I, III, IV, and V in hypertension. Interestingly, targeted examination of the brain stem, a regulator of cardiovascular homeostasis and systemic blood pressure, further showed the occurrence of mitochondrial complex I dysfunction, elevated reactive oxygen species production, decreased ATP synthesis, and impaired respiration in hypertension. Our findings suggest that in already-hypertensive spontaneously hypertensive rats, the brain respiratory complexes exhibit previously unknown assembly defects. These defects impair the function of the mitochondrial respiratory chain. This mitochondrial dysfunction localizes to the brain stem and is, therefore, likely to contribute to the development, as well as to pathophysiological complications, of hypertension.


Subject(s)
Brain/enzymology , Hypertension/complications , Hypertension/physiopathology , Mitochondrial Diseases/etiology , Multienzyme Complexes/genetics , Protein Processing, Post-Translational , Animals , Cardiovascular System/physiopathology , Doxycycline/pharmacology , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/pharmacology , Homeostasis , Hypertension/enzymology , Metalloendopeptidases/antagonists & inhibitors , Mitochondrial Diseases/enzymology , Multienzyme Complexes/drug effects , Protein Processing, Post-Translational/drug effects , Proteomics , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Tandem Mass Spectrometry
17.
Mol Cell Proteomics ; 4(8): 1205-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15911532

ABSTRACT

Here we describe a proteomic analysis of Escherichia coli in which 3,199 protein forms were detected, and of those 2,160 were annotated and assigned to the cytosol, periplasm, inner membrane, and outer membrane by biochemical fractionation followed by two-dimensional gel electrophoresis and tandem mass spectrometry. Represented within this inventory were unique and modified forms corresponding to 575 different ORFs that included 151 proteins whose existence had been predicted from hypothetical ORFs, 76 proteins of completely unknown function, and 222 proteins currently without location assignments in the Swiss-Prot Database. Of the 575 unique proteins identified, 42% were found to exist in multiple forms. Using DIGE, we also examined the relative changes in protein expression when cells were grown in the presence and absence of amino acids. A total of 23 different proteins were identified whose abundance changed significantly between the two conditions. Most of these changes were found to be associated with proteins involved in carbon and amino acid metabolism, transport, and chemotaxis. Detailed information related to all 2,160 protein forms (protein and gene names, accession numbers, subcellular locations, relative abundances, sequence coverage, molecular masses, and isoelectric points) can be obtained upon request in either tabular form or as interactive gel images.


Subject(s)
Amino Acids/deficiency , Bacterial Proteins/chemistry , Escherichia coli/chemistry , Escherichia coli/growth & development , Genome, Bacterial , Bacterial Proteins/isolation & purification , Databases, Protein , Electrophoresis, Gel, Two-Dimensional/methods , Open Reading Frames , Peptide Mapping/methods , Sequence Analysis/methods
18.
Circ Res ; 94(1): 68-76, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14656925

ABSTRACT

Matrix metalloproteinase (MMP)-dependent shedding of heparin-binding epidermal growth factor (HB-EGF) and subsequent activation of the EGF receptor (EGFR) in the cardiovasculature is emerging as a unique mechanism signaling growth effects of diverse G protein-coupled receptors (GPCRs). Among these GPCRs are adrenoceptors and angiotensin receptors that contribute to the pathogenesis of hypertension through their vasoconstrictive and growth effects. Focusing on alpha(1b)-adrenoceptors, we suggest here that MMP-dependent activation of the EGFR promotes vasoconstriction as well as growth. We identified MMP-7 as a major HB-EGF sheddase in rat mesenteric arteries and alpha(1b)-adrenoceptors, angiotensin receptors, and hypertension-stimulated MMP-7 activity. Adrenoceptors stimulated EGFR autophosphorylation in arteries, and this transactivation was opposed by the MMP-7 inhibitor GM6001 as well as MMP-7-specific antibodies. In isolated microperfused arteries, blockade of EGFR transactivation with inhibitors of the EGFR (AG1478 and PD153035), HB-EGF (CRM197 and neutralizing antibodies), or MMPs (doxycycline) inhibited adrenergic vasoconstriction. In spontaneously hypertensive rats but not in normotensive rats, the inhibition of MMPs with doxycycline (19.2 mg/d from week 7 until week 12) reduced systolic blood pressure and attenuated HB-EGF shedding in the mesenteric arteries. These findings suggest a previously unknown mechanism of vasoregulation whereby agonists of certain GPCRs (such as adrenoceptors and angiotensin receptors) activate MMPs (such as MMP-7) that shed EGFR ligands (such as HB-EGF), which then activate the EGFR, thereby promoting vasoconstriction as well as growth. Because this mechanism is triggered by agonists typically overexpressed in hypertension, its blockade may have therapeutic potential for simultaneously inhibiting pathological vasoconstriction and growth in hypertensive disorders.


Subject(s)
ErbB Receptors/metabolism , Hypertension/physiopathology , Matrix Metalloproteinase 7/metabolism , Vasoconstriction , Adrenergic alpha-1 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Animals , Arteries/enzymology , Arteries/physiology , Blood Pressure , Enzyme Activation , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Heparin-binding EGF-like Growth Factor , Hypertension/enzymology , Intercellular Signaling Peptides and Proteins , Male , Phenylephrine/pharmacology , Rats , Rats, Inbred WKY , Rats, Sprague-Dawley , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL