Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Nanotechnol ; 19(1): 106-114, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37709951

ABSTRACT

Quantum biological tunnelling for electron transfer is involved in controlling essential functions for life such as cellular respiration and homoeostasis. Understanding and controlling the quantum effects in biology has the potential to modulate biological functions. Here we merge wireless nano-electrochemical tools with cancer cells for control over electron transfer to trigger cancer cell death. Gold bipolar nanoelectrodes functionalized with redox-active cytochrome c and a redox mediator zinc porphyrin are developed as electric-field-stimulating bio-actuators, termed bio-nanoantennae. We show that a remote electrical input regulates electron transport between these redox molecules, which results in quantum biological tunnelling for electron transfer to trigger apoptosis in patient-derived cancer cells in a selective manner. Transcriptomics data show that the electric-field-induced bio-nanoantenna targets the cancer cells in a unique manner, representing electrically induced control of molecular signalling. The work shows the potential of quantum-based medical diagnostics and treatments.


Subject(s)
Apoptosis , Neoplasms , Humans , Electron Transport , Oxidation-Reduction , Cell Death , Gold/chemistry
2.
Arterioscler Thromb Vasc Biol ; 43(3): 456-473, 2023 03.
Article in English | MEDLINE | ID: mdl-36700427

ABSTRACT

BACKGROUND: Late vein graft failure is caused by intimal thickening resulting from endothelial cell (EC) damage and inflammation which promotes vascular smooth muscle cell (VSMC) dedifferentiation, migration, and proliferation. Nonphosphorylatable PRH (proline-rich homeodomain) S163C:S177C offers enhanced stability and sustained antimitotic effect. Therefore, we investigated whether adenovirus-delivered PRH S163C:S177C protein attenuates intimal thickening via VSMC phenotype modification without detrimental effects on ECs. METHODS: PRH S163C:S177C was expressed in vitro (human saphenous vein-VSMCs and human saphenous vein-ECs) and in vivo (ligated mouse carotid arteries) by adenoviruses. Proliferation, migration, and apoptosis were quantified and phenotype was assessed using Western blotting for contractile filament proteins and collagen gel contraction. EC inflammation was quantified using VCAM (vascular cell adhesion protein)-1, ICAM (intercellular adhesion molecule)-1, interleukin-6, and monocyte chemotactic factor-1 measurement and monocyte adhesion. Next Generation Sequencing was utilized to identify novel downstream mediators of PRH action and these and intimal thickening were investigated in vivo. RESULTS: PRH S163C:S177C inhibited proliferation, migration, and apoptosis and promoted contractile phenotype (enhanced contractile filament proteins and collagen gel contraction) compared with virus control in human saphenous vein-VSMCs. PRH S163C:S177C expression in human saphenous vein-ECs significantly reduced apoptosis, without affecting cell proliferation and migration, while reducing TNF (tumor necrosis factor)-α-induced VCAM-1 and ICAM-1 and monocyte adhesion and suppressing interleukin-6 and monocyte chemotactic factor-1 protein levels. PRH S163C:S177C expression in ligated murine carotid arteries significantly impaired carotid artery ligation-induced neointimal proliferation and thickening without reducing endothelial coverage. Next Generation Sequencing revealed STAT-1 (signal transducer and activator of transcription 1) and HDAC-9 (histone deacetylase 9) as mediators of PRH action and was supported by in vitro and in vivo analyses. CONCLUSIONS: We observed PRH S163C:S177C attenuated VSMC proliferation, and migration and enhanced VSMC differentiation at least in part via STAT-1 and HDAC-9 signaling while promoting endothelial repair and anti-inflammatory properties. These findings highlight the potential for PRH S163C:S177C to preserve endothelial function whilst suppressing intimal thickening, and reducing late vein graft failure.


Subject(s)
Interleukin-6 , Tunica Intima , Mice , Animals , Humans , Interleukin-6/metabolism , Tunica Intima/pathology , Cell Proliferation , Neointima/pathology , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Myocytes, Smooth Muscle/metabolism , Cell Movement
3.
Methods Mol Biol ; 2441: 257-275, 2022.
Article in English | MEDLINE | ID: mdl-35099743

ABSTRACT

Interactions between DNA and proteins are crucial for the regulation of gene expression. Chromatin immunoprecipitation (ChIP) is a powerful technique that allows the study of specific protein-DNA interactions in cultured cells and fresh or fixed tissue. Chromatin is isolated and sheared, and antibodies against the protein(s) of interest are used to isolate specific protein-DNA complexes. Subsequent analysis by real-time polymerase chain reaction (qPCR) or next-generation sequencing (NGS) allows identification and quantification of the co-purified DNA fragments, and NGS also gives insight into the genomic binding sites of a protein. Here we describe a cross-linking ChIP (X-ChIP) protocol, based around the example of a myc-tagged Proline-Rich Homeodomain (PRH) protein expressed in human umbilical vein endothelial cells. We also describe how to analyse specific known or suspected binding sites using quantitative PCR as well as how to analyse genome-wide binding from ChIP sequencing data.


Subject(s)
Endothelial Cells , Transcription Factors , Binding Sites , Chromatin/genetics , Chromatin Immunoprecipitation/methods , Endothelial Cells/metabolism , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Anticancer Res ; 41(12): 6155-6167, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34848470

ABSTRACT

BACKGROUND: The incidence of cholangiocarcinoma (CCA) is increasing worldwide and current single chemotherapeutic drug treatments are ineffective. CX-4945 and cisplatin are currently in clinical trial for CCA treatment. MATERIALS AND METHODS: We assessed the effects of the sequence of administration of CX-4945 and cisplatin applied in combination treatments on their efficacy in CCA cells in vitro. CCA cell viability was examined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Apoptosis was examined using flow cytometry. The percentage of cells positive for phosphorylated H2A histone family member X (γ-H2AX) were measured using both flow cytometry and immunofluorescence. RESULTS: CCA cell viability was reduced to 50% after 24 h of treatments with CX-4945 and cisplatin as single agents. Interestingly, treatment with cisplatin 6 h prior to CX-4945 treatment induced significantly more DNA damage and apoptosis than CX-4945 treatment followed by cisplatin. Unexpectedly, CX-4945 treatment followed by cisplatin was less effective than single treatment in RMCCA-1 CCA cells. In addition, a 1:1 ratio of each drug was the most effective combination in these cells. CONCLUSION: These data demonstrate that the combination of CX-4945 and cis platin acts additively when cisplatin is applied first, at least in part due to increased DNA damage and apoptosis. Furthermore, treatment with CX-4945 prior to cisplatin treatment reduces the efficacy of this drug combination in CCA cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Cholangiocarcinoma/drug therapy , Cisplatin/therapeutic use , Naphthyridines/therapeutic use , Phenazines/therapeutic use , Antineoplastic Agents/pharmacology , Cell Proliferation , Cisplatin/pharmacology , Drug Combinations , Drug Synergism , Humans , Naphthyridines/pharmacology , Phenazines/pharmacology
6.
Explor Target Antitumor Ther ; 2(5): 434-447, 2021.
Article in English | MEDLINE | ID: mdl-36045705

ABSTRACT

Cholangiocarcinoma (CCA) is a disease with a very poor prognosis and limited treatment options. Although targeted therapies directed towards specific mutations found in CCA are becoming available and are showing great potential, many tumors do not carry actionable mutations and, in those that do, the emergence of drug resistance is a likely consequence of treatment. Therapeutic targeting of enzymes and other proteins that show elevated activity in CCA cells but which are not altered by mutation is a potential strategy for the treatment of target negative and drug-resistant disease. Protein kinase CK2 (CK2) is a ubiquitously expressed kinase that has increased expression and increased activity in a variety of cancer types including CCA. Several potent CK2 inhibitors are in pre-clinical development or under assessment in a variety of clinical trials often in combination with drugs that induce DNA damage. This review outlines the importance of CK2 in CCA and assesses the progress that has been made in the evaluation of CK2 inhibition as a treatment strategy in this disease. Targeting CK2 based on the expression levels or activity of this protein and/or in combination with drugs that induce DNA damage or inhibit cell cycle progression, could be a viable option for tumors that lack actionable mutations, or for tumors that develop resistance to targeted treatments.

7.
Oncogenesis ; 9(2): 10, 2020 Feb 04.
Article in English | MEDLINE | ID: mdl-32019914

ABSTRACT

Cancer cells go through a process known as epithelial-mesenchymal transition (EMT) during which they acquire the ability to migrate and invade extracellular matrix. Some cells also acquire the ability to move across a layer of endothelial cells to enter and exit the bloodstream; intra- and extravasation, respectively. The transcription factor PRH/HHEX (proline-rich homeodomain/haematopoietically expressed homeobox) controls cell proliferation and cell migration/invasion in a range of cell types. Our previous work showed that PRH activity is downregulated in prostate cancer cells owing to increased inhibitory PRH phosphorylation and that this increases cell proliferation and invasion. PRH inhibits migration and invasion by prostate and breast epithelial cells in part by activating the transcription of Endoglin, a transforming growth factor ß (TGFß) co-receptor. Here we show that depletion of PRH in immortalised prostate epithelial cells results in increased extravasation in vitro. We show that blood platelets stimulate extravasation of cells with depleted PRH and that inhibition of TGFß signalling blocks the effects of platelets on these cells. Moreover, TGFß induces changes characteristic of EMT including decreased E-Cadherin expression and increased Snail expression. We show that in prostate cells PRH regulates multiple genes involved in EMT and TGFß signalling. However, both platelets and TGFß increase PRH phosphorylation. In addition, TGFß increases binding of its effector pSMAD3 to the PRH/HHEX promoter and downregulates PRH protein and mRNA levels. Thus, TGFß signalling downregulates PRH activity by multiple mechanisms and induces an EMT that facilitates extravasation and sensitises cells to TGFß.

8.
Cancer Res ; 80(4): 757-770, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31843982

ABSTRACT

Aberrant Notch and Wnt signaling are known drivers of cholangiocarcinoma (CCA), but the underlying factors that initiate and maintain these pathways are not known. Here, we show that the proline-rich homeodomain protein/hematopoietically expressed homeobox (PRH/HHEX) transcription factor forms a positive transcriptional feedback loop with Notch3 that is critical in CCA. PRH/HHEX expression is elevated in CCA, and depletion of PRH reduces CCA tumor growth in a xenograft model. Overexpression of PRH in primary human biliary epithelial cells is sufficient to increase cell proliferation and produce an invasive phenotype. Interrogation of the gene networks regulated by PRH and Notch3 reveals that unlike Notch3, PRH directly activates canonical Wnt signaling. These data indicate that hyperactivation of Notch and Wnt signaling is independent of the underlying mutational landscape and has a common origin in dysregulation of PRH. Moreover, they suggest new therapeutic options based on the dependence of specific Wnt, Notch, and CDK4/6 inhibitors on PRH activity. SIGNIFICANCE: The PRH/HHEX transcription factor is an oncogenic driver in cholangiocarcinoma that confers sensitivity to CDK4/6 inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Homeodomain Proteins/metabolism , Receptor, Notch3/metabolism , Transcription Factors/metabolism , Animals , Antineoplastic Agents/therapeutic use , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Epithelial Cells , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Regulatory Networks , Homeodomain Proteins/genetics , Humans , K562 Cells , Male , Mice , Mutation , Neoplasm Invasiveness/genetics , Piperazines/pharmacology , Piperazines/therapeutic use , Primary Cell Culture , Promoter Regions, Genetic , Pyridines/pharmacology , Pyridines/therapeutic use , RNA-Seq , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
9.
Cancers (Basel) ; 10(9)2018 Aug 23.
Article in English | MEDLINE | ID: mdl-30142881

ABSTRACT

Cholangiocarcinoma is a disease with a poor prognosis and increasing incidence and hence there is a pressing unmet clinical need for new adjuvant treatments. Protein kinase CK2 (previously casein kinase II) is a ubiquitously expressed protein kinase that is up-regulated in multiple cancer cell types. The inhibition of CK2 activity using CX-4945 (Silmitasertib) has been proposed as a novel treatment in multiple disease settings including cholangiocarcinoma. Here, we show that CX-4945 inhibited the proliferation of cholangiocarcinoma cell lines in vitro. Moreover, CX-4945 treatment induced the formation of cytosolic vacuoles in cholangiocarcinoma cell lines and other cancer cell lines. The vacuoles contained extracellular fluid and had neutral pH, features characteristic of methuosis. In contrast, simultaneous knockdown of both the α and α' catalytic subunits of protein kinase CK2 using small interfering RNA (siRNA) had little or no effect on the proliferation of cholangiocarcinoma cell lines and failed to induce the vacuole formation. Surprisingly, low doses of CX-4945 increased the invasive properties of cholangiocarcinoma cells due to an upregulation of matrix metallopeptidase 7 (MMP-7), while the knockdown of CK2 inhibited cell invasion. Our data suggest that CX-4945 inhibits cell proliferation and induces cell death via CK2-independent pathways. Moreover, the increase in cell invasion brought about by CX-4945 treatment suggests that this drug might increase tumor invasion in clinical settings.

10.
Nat Commun ; 9(1): 2704, 2018 07 13.
Article in English | MEDLINE | ID: mdl-30006544

ABSTRACT

Formation of the lymphatic system requires the coordinated expression of several key regulators: vascular endothelial growth factor C (VEGFC), its receptor FLT4, and a key transcriptional effector, PROX1. Yet, how expression of these signaling components is regulated remains poorly understood. Here, using a combination of genetic and molecular approaches, we identify the transcription factor hematopoietically expressed homeobox (HHEX) as an upstream regulator of VEGFC, FLT4, and PROX1 during angiogenic sprouting and lymphatic formation in vertebrates. By analyzing zebrafish mutants, we found that hhex is necessary for sprouting angiogenesis from the posterior cardinal vein, a process required for lymphangiogenesis. Furthermore, studies of mammalian HHEX using tissue-specific genetic deletions in mouse and knockdowns in cultured human endothelial cells reveal its highly conserved function during vascular and lymphatic development. Our findings that HHEX is essential for the regulation of the VEGFC/FLT4/PROX1 axis provide insights into the molecular regulation of lymphangiogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Lymphangiogenesis/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Base Sequence , Blood Vessels/cytology , Blood Vessels/growth & development , Blood Vessels/metabolism , Cell Line , Embryo, Mammalian , Embryo, Nonmammalian , Endothelial Cells/cytology , Endothelial Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mice , Neovascularization, Physiologic/genetics , Repressor Proteins/deficiency , Signal Transduction , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism
11.
J Biol Chem ; 291(29): 15029-45, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27151212

ABSTRACT

Urokinase-type plasminogen activator (uPA) regulates angiogenesis and vascular permeability through proteolytic degradation of extracellular matrix and intracellular signaling initiated upon its binding to uPAR/CD87 and other cell surface receptors. Here, we describe an additional mechanism by which uPA regulates angiogenesis. Ex vivo VEGF-induced vascular sprouting from Matrigel-embedded aortic rings isolated from uPA knock-out (uPA(-/-)) mice was impaired compared with vessels emanating from wild-type mice. Endothelial cells isolated from uPA(-/-) mice show less proliferation and migration in response to VEGF than their wild type counterparts or uPA(-/-) endothelial cells in which expression of wild type uPA had been restored. We reported previously that uPA is transported from cell surface receptors to nuclei through a mechanism that requires its kringle domain. Intranuclear uPA modulates gene transcription by binding to a subset of transcription factors. Here we report that wild type single-chain uPA, but not uPA variants incapable of nuclear transport, increases the expression of cell surface VEGF receptor 1 (VEGFR1) and VEGF receptor 2 (VEGFR2) by translocating to the nuclei of ECs. Intranuclear single-chain uPA binds directly to and interferes with the function of the transcription factor hematopoietically expressed homeodomain protein or proline-rich homeodomain protein (HHEX/PRH), which thereby lose their physiologic capacity to repress the activity of vehgr1 and vegfr2 gene promoters. These studies identify uPA-dependent de-repression of vegfr1 and vegfr2 gene transcription through binding to HHEX/PRH as a novel mechanism by which uPA mediates the pro-angiogenic effects of VEGF and identifies a potential new target for control of pathologic angiogenesis.


Subject(s)
Homeodomain Proteins/metabolism , Neovascularization, Physiologic , Transcription Factors/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , HEK293 Cells , Humans , K562 Cells , Mice, Knockout , Neovascularization, Physiologic/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
12.
Cell Biosci ; 6: 12, 2016.
Article in English | MEDLINE | ID: mdl-26877867

ABSTRACT

The proline rich homeodomain protein (PRH), also known as haematopoietically expressed homeobox (HHEX), is an essential transcription factor in embryonic development and in the adult. The PRH protein forms oligomeric complexes that bind to tandemly repeated PRH recognition sequences within or at a distance from PRH-target genes and recruit a variety of PRH-interacting proteins. PRH can also bind to other transcription factors and co-regulate specific target genes either directly through DNA binding, or indirectly through effects on the activity of its partner proteins. In addition, like some other homeodomain proteins, PRH can regulate the translation of specific mRNAs. Altered PRH expression and altered PRH intracellular localisation, are associated with breast cancer, liver cancer and thyroid cancer and some subtypes of leukaemia. This is consistent with the involvement of multiple PRH-interacting proteins, including the oncoprotein c-Myc, translation initiation factor 4E (eIF4E), and the promyelocytic leukaemia protein (PML), in the control of cell proliferation and cell survival. Similarly, multiple PRH target genes, including the genes encoding vascular endothelial growth factor (VEGF), VEGF receptors, Endoglin, and Goosecoid, are known to be important in the control of cell proliferation and cell survival and/or the regulation of cell migration and invasion. In this review, we summarise the evidence that implicates PRH in tumourigenesis and we review the data that suggests PRH levels could be useful in cancer prognosis and in the choice of treatment options.

13.
Leuk Res ; 36(11): 1434-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22874537

ABSTRACT

The PRH/Hhex transcription factor represses multiple genes in the VEGF signalling pathway (VSP) to inhibit myeloid cell survival. Protein kinase CK2 phosphorylates PRH and counteracts the inhibitory effect of this protein on cell survival by blocking the repression of VSP genes. Here we show that the BCR-ABL/Src kinase inhibitor dasatinib decreases PRH phosphorylation and increases PRH-dependent repression of Vegf and Vegfr-1. Moreover in the absence of PRH, dasatinib does not inhibit cell survival as effectively as in PRH expressing cells. Thus the re-establishment of gene control by PRH is in part responsible for the therapeutic effects of dasatinib.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/metabolism , Leukemia/metabolism , Pyrimidines/pharmacology , Signal Transduction , Thiazoles/pharmacology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Dasatinib , Humans , Phosphorylation/drug effects , Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/genetics
14.
Nucleic Acids Res ; 40(18): 9008-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22844093

ABSTRACT

Protein kinase CK2 promotes cell survival and the activity of this kinase is elevated in several cancers including chronic myeloid leukaemia. We have shown previously that phosphorylation of the Proline-Rich Homeodomain protein (PRH/Hhex) by CK2 inhibits the DNA-binding activity of this transcription factor. Furthermore, PRH represses the transcription of multiple genes encoding components of the VEGF-signalling pathway and thereby influences cell survival. Here we show that the inhibitory effects of PRH on cell proliferation are abrogated by CK2 and that CK2 inhibits the binding of PRH at the Vegfr-1 promoter. Phosphorylation of PRH by CK2 also decreases the nuclear association of PRH and induces its cleavage by the proteasome. Moreover, cleavage of phosphorylated PRH produces a stable truncated cleavage product which we have termed PRHΔC (HhexΔC). PRHΔC acts as a transdominant negative regulator of full-length PRH by sequestering TLE proteins that function as PRH co-repressors. We show that this novel regulatory mechanism results in the alleviation of PRH-mediated repression of Vegfr-1. We suggest that the re-establishment of PRH function through inhibition of CK2 could be of value in treatment of myeloid leukaemias, as well as other tumour types in which PRH is inactivated by phosphorylation.


Subject(s)
Casein Kinase II/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Cell Proliferation , Cell Survival , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , K562 Cells , Promoter Regions, Genetic , Protein Stability , Repressor Proteins/metabolism , Sequence Deletion , Signal Transduction/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/physiology
15.
PLoS One ; 7(4): e35984, 2012.
Article in English | MEDLINE | ID: mdl-22540015

ABSTRACT

BACKGROUND: Many transcription factors control gene expression by binding to specific DNA sequences at or near the genes that they regulate. However, some transcription factors play more global roles in the control of gene expression by altering the architecture of sections of chromatin or even the whole genome. The ability to form oligomeric protein assemblies allows many of these proteins to manipulate extensive segments of DNA or chromatin via the formation of structures such as DNA loops or protein-DNA fibres. PRINCIPAL FINDINGS: Here we show that the proline rich homeodomain protein PRH/Hhex forms predominantly octameric and/or hexadecameric species in solution as well as larger assemblies. We show that these assemblies are highly stable resisting denaturation by temperature and chemical denaturants. CONCLUSION: These data indicate that PRH is functionally and structurally related to the Lrp/AsnC family of proteins, a group of proteins that are known to act globally to control gene expression in bacteria and archaea.


Subject(s)
Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Circular Dichroism , Guanidines/chemistry , Homeodomain Proteins/chemistry , Protein Denaturation , Protein Multimerization , Protein Structure, Secondary , Temperature , Thiocyanates/chemistry , Transcription Factors/chemistry
16.
J Vis Exp ; (41)2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20689508

ABSTRACT

Protein function is intimately coupled to protein localization. Although some proteins are restricted to a specific location or subcellular compartment, many proteins are present as a freely diffusing population in free exchange with a sub-population that is tightly associated with a particular subcellular domain or structure. In situ subcellular fractionation allows the visualization of protein compartmentalization and can also reveal protein sub-populations that localize to specific structures. For example, removal of soluble cytoplasmic proteins and loosely held nuclear proteins can reveal the stable association of some transcription factors with chromatin. Subsequent digestion of DNA can in some cases reveal association with the network of proteins and RNAs that is collectively termed the nuclear scaffold or nuclear matrix. Here we describe the steps required during the in situ fractionation of adherent and non-adherent mammalian cells on microscope coverslips. Protein visualization can be achieved using specific antibodies or fluorescent fusion proteins and fluorescence microscopy. Antibodies and/or fluorescent dyes that act as markers for specific compartments or structures allow protein localization to be mapped in detail. In situ fractionation can also be combined with western blotting to compare the amounts of protein present in each fraction. This simple biochemical approach can reveal associations that would otherwise remain undetected.


Subject(s)
Cell Adhesion/physiology , Cell Fractionation/methods , Subcellular Fractions/chemistry , Animals , COS Cells , Chlorocebus aethiops , Humans , Mammals , Polylysine/chemistry
17.
Nucleic Acids Res ; 38(21): 7513-25, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20675722

ABSTRACT

Protein self-organization is essential for the establishment and maintenance of nuclear architecture and for the regulation of gene expression. We have shown previously that the Proline-Rich Homeodomain protein (PRH/Hex) self-assembles to form oligomeric complexes that bind to arrays of PRH binding sites with high affinity and specificity. We have also shown that many PRH target genes contain suitably spaced arrays of PRH sites that allow this protein to bind and regulate transcription. Here, we use analytical ultracentrifugation and electron microscopy to further characterize PRH oligomers. We use the same techniques to show that PRH oligomers bound to long DNA fragments self-associate to form highly ordered assemblies. Electron microscopy and linear dichroism reveal that PRH oligomers can form protein-DNA fibres and that PRH is able to compact DNA in the absence of other proteins. Finally, we show that DNA compaction is not sufficient for the repression of PRH target genes in cells. We conclude that DNA compaction is a consequence of the binding of large PRH oligomers to arrays of binding sites and that PRH is functionally and structurally related to the Lrp/AsnC family of proteins from bacteria and archaea, a group of proteins formerly thought to be without eukaryotic equivalents.


Subject(s)
DNA/chemistry , Homeodomain Proteins/chemistry , Transcription Factors/chemistry , Binding Sites , DNA/metabolism , DNA/ultrastructure , Homeodomain Proteins/metabolism , Homeodomain Proteins/ultrastructure , Humans , K562 Cells , Nucleic Acid Conformation , Protein Multimerization , Transcription Factors/metabolism , Transcription Factors/ultrastructure , Transcription, Genetic
18.
Hypertension ; 55(3): 689-97, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20124108

ABSTRACT

The proteinase-activated receptor 2 (PAR-2) expression is increased in endothelial cells derived from women with preeclampsia, characterized by widespread maternal endothelial damage, which occurs as a consequence of elevated soluble vascular endothelial growth factor receptor-1 (sVEGFR-1; commonly known as sFlt-1) in the maternal circulation. Because PAR-2 is upregulated by proinflammatory cytokines and activated by blood coagulation serine proteinases, we investigated whether activation of PAR-2 contributed to sVEGFR-1 release. PAR-2-activating peptides (SLIGRL-NH(2) and 2-furoyl-LIGRLO-NH(2)) and factor Xa increased the expression and release of sVEGFR-1 from human umbilical vein endothelial cells. Enzyme-specific, dominant-negative mutants and small interfering RNA were used to demonstrate that PAR-2-mediated sVEGFR-1 release depended on protein kinase C-beta(1) and protein kinase C-epsilon, which required intracellular transactivation of epidermal growth factor receptor 1, leading to mitogen-activated protein kinase activation. Overexpression of heme oxygenase 1 and its gaseous product, carbon monoxide, decreased PAR-2-stimulated sVEGFR-1 release from human umbilical vein endothelial cells. Simvastatin, which upregulates heme oxygenase 1, also suppressed PAR-2-mediated sVEGFR-1 release. These results show that endothelial PAR-2 activation leading to increased sVEGFR-1 release may contribute to the maternal vascular dysfunction observed in preeclampsia and highlights the PAR-2 pathway as a potential therapeutic target for the treatment of preeclampsia.


Subject(s)
Endothelial Cells/metabolism , ErbB Receptors/metabolism , Pre-Eclampsia/metabolism , Receptor, PAR-2/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Carbon Monoxide/metabolism , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/physiology , Endothelial Cells/cytology , Factor Xa/metabolism , Female , Gene Expression/physiology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney/cytology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Pregnancy , Protein Kinase C/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor, PAR-2/agonists , Simvastatin/pharmacology , Solubility , Umbilical Veins/cytology , Vascular Endothelial Growth Factor Receptor-1/genetics
19.
Mol Cell Biol ; 30(9): 2120-34, 2010 May.
Article in English | MEDLINE | ID: mdl-20176809

ABSTRACT

The proline-rich homeodomain protein (PRH) plays multiple roles in the control of gene expression during embryonic development and in the adult. Vascular endothelial growth factor (VEGF) is a mitogen that stimulates cell proliferation and survival via cell surface receptors including VEGFR-1 and VEGFR-2. VEGF signaling is of critical importance in angiogenesis and hematopoiesis and is elevated in many tumors. Here we show that PRH binds directly to the promoter regions of the Vegf, Vegfr-1, and Vegfr-2 genes and that in each case PRH represses transcription. We demonstrate that overexpression or knockdown of PRH directly impinges on the survival of both leukemic and tumor cells and that the modulation of VEGF and VEGF receptor signaling by PRH mediates these effects. Our findings demonstrate that PRH is a key regulator of the VEGF signaling pathway and describe a mechanism whereby PRH plays an important role in tumorigenesis and leukemogenesis.


Subject(s)
Homeodomain Proteins/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Transcription, Genetic , Vascular Endothelial Growth Factor A/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Death , Cell Proliferation , Cell Survival , Co-Repressor Proteins , DNA, Neoplasm/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , K562 Cells , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
Nucleic Acids Res ; 37(10): 3288-300, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19324893

ABSTRACT

The proline-rich homeodomain protein (PRH/Hex) regulates transcription by binding to specific DNA sequences and regulates mRNA transport by binding to translation initiation factor eIF4E. Protein kinase CK2 plays multiple roles in the regulation of gene expression and cell proliferation. Here, we show that PRH interacts with the beta subunit of CK2 in vitro and in cells and that CK2 phosphorylates PRH. Phosphorylation of PRH by CK2 inhibits the DNA binding activity of this protein and dephosphorylation restores DNA binding indicating that this modification acts as a reversible switch. We show that phosphorylation of the homeodomain is sufficient to block DNA binding and we identify two amino acids within this the domain that are phosphorylated by CK2: S163 and S177. Site-directed mutagenesis demonstrates that mutation of either of these residues to glutamic acid partially mimics phosphorylation but is insufficient to completely block DNA binding whereas an S163E/S177E double mutation severely inhibits DNA binding. Significantly, the S163E and S177E mutations and the S163E/S177E double mutation all inhibit the ability of PRH to regulate transcription in cells. Since these amino acids are conserved between many homeodomain proteins, our results suggest that CK2 may regulate the activity of several homeodomain proteins in this manner.


Subject(s)
Casein Kinase II/metabolism , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , DNA/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/chemistry , Humans , K562 Cells , Molecular Sequence Data , Mutation , Phosphoproteins/metabolism , Phosphorylation , Protein Structure, Tertiary , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Serine/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...