Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 37(30): 4110-4121, 2018 07.
Article in English | MEDLINE | ID: mdl-29706656

ABSTRACT

Glioblastoma (GBM) is the most aggressive and an incurable type of brain cancer. Human cytomegalovirus (HCMV) DNA and encoded proteins, including the chemokine receptor US28, have been detected in GBM tumors. US28 displays constitutive activity and is able to bind several human chemokines, leading to the activation of various proliferative and inflammatory signaling pathways. Here we show that HCMV, through the expression of US28, significantly enhanced the growth of 3D spheroids of U251- and neurospheres of primary glioblastoma cells. Moreover, US28 expression accelerated the growth of glioblastoma cells in an orthotopic intracranial GBM-model in mice. We developed highly potent and selective US28-targeting nanobodies, which bind to the extracellular domain of US28 and detect US28 in GBM tissue. The nanobodies inhibited chemokine binding and reduced the constitutive US28-mediated signaling with nanomolar potencies and significantly impaired HCMV/US28-mediated tumor growth in vitro and in vivo. This study emphasizes the oncomodulatory role of HCMV-encoded US28 and provides a potential therapeutic approach for HCMV-positive tumors using the nanobody technology.


Subject(s)
Brain Neoplasms/genetics , Cell Proliferation/genetics , Cytomegalovirus/genetics , Glioblastoma/genetics , Receptors, Chemokine/genetics , Viral Proteins/genetics , Animals , Brain Neoplasms/pathology , COS Cells , Cell Line , Chlorocebus aethiops , Female , Glioblastoma/pathology , HEK293 Cells , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Receptors, Virus/genetics , Signal Transduction/genetics
2.
J Pharmacol Exp Ther ; 363(1): 35-44, 2017 10.
Article in English | MEDLINE | ID: mdl-28768817

ABSTRACT

WHIM syndrome is a rare congenital immunodeficiency disease, named after its main clinical manifestations: warts, hypogammaglobulinemia, infections, and myelokathexis, which refers to abnormal accumulation of mature neutrophils in the bone marrow. The disease is primarily caused by C-terminal truncation mutations of the chemokine receptor CXCR4, giving these CXCR4-WHIM mutants a gain of function in response to their ligand CXCL12. Considering the broad functions of CXCR4 in maintaining leukocyte homeostasis, patients are panleukopenic and display altered immune responses, likely as a consequence of impairment in the differentiation and trafficking of leukocytes. Treatment of WHIM patients currently consists of symptom relief, leading to unsatisfactory clinical responses. As an alternative and potentially more effective approach, we tested the potency and efficacy of CXCR4-specific nanobodies on inhibiting CXCR4-WHIM mutants. Nanobodies are therapeutic proteins based on the smallest functional fragments of heavy chain antibodies. They combine the advantages of small-molecule drugs and antibody-based therapeutics due to their relative small size, high stability, and high affinity. We compared the potential of monovalent and bivalent CXCR4-specific nanobodies to inhibit CXCL12-induced CXCR4-WHIM-mediated signaling with the small-molecule clinical candidate AMD3100. The CXCR4-targeting nanobodies displace CXCL12 binding and bind CXCR4-wild type and CXCR4-WHIM (R334X/S338X) mutants and with (sub-) nanomolar affinities. The nanobodies' epitope was mapped to extracellular loop 2 of CXCR4, overlapping with the binding site of CXCL12. Monovalent, and in particular bivalent, nanobodies were more potent than AMD3100 in reducing CXCL12-mediated G protein activation. In addition, CXCR4-WHIM-dependent calcium flux and wound healing of human papillomavirus-immortalized cell lines in response to CXCL12 was effectively inhibited by the nanobodies. Based on these in vitro results, we conclude that CXCR4 nanobodies hold significant potential as alternative therapeutics for CXCR4-associated diseases such as WHIM syndrome.


Subject(s)
Antibody Specificity , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/therapy , Receptors, CXCR4/immunology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/therapeutic use , Warts/immunology , Warts/therapy , HEK293 Cells , Humans , Immunologic Deficiency Syndromes/genetics , Mutation , Primary Immunodeficiency Diseases , Receptors, CXCR4/genetics , Warts/genetics
3.
Transplantation ; 101(3): 531-540, 2017 03.
Article in English | MEDLINE | ID: mdl-27362315

ABSTRACT

BACKGROUND: Renal transplantation is the preferred treatment for patients with end-stage renal disease. Human cytomegalovirus (HCMV) activation is associated with decreased renal graft function and survival. Human cytomegalovirus encodes several immune modulatory proteins, including the G protein-coupled receptor US28, which scavenges human chemokines and modulates intracellular signaling. METHODS: Our aim was to identify the expression and localization of US28 in renal allograft biopsies by immunohistochemistry and determine its role in viral spreading in vitro. RESULTS: Immunohistochemistry revealed US28 in 31 of 34 renal transplant biopsies from HCMV-seropositive donors. Expression was independent of HCMV viremia or IgG serostatus. US28 was predominantly expressed in the cytoplasm of vascular smooth muscle cells (VSMCs) and tubular epithelial cells, with a median positivity of 20% and 40%, respectively. Also, US28-positive cells were present within arterial neointima. In contrast to US28, HCMV-encoded immediate early antigen was detected in less than 5% of VSMCs, tubular epithelial cells, interstitial endothelium, interstitial inflammatory infiltrates, and glomerular cells.Primary VSMCs were infected with green fluorescent protein-tagged wild type or US28-deficient HCMV. The viral spreading of US28-deficient HCMV, via culture medium or cell-to-cell transmission, was significantly impeded as shown by green fluorescent protein (ie, infected) cell quantification and quantitative real-time polymerase chain reaction. Additionally, the number and size of foci was smaller. CONCLUSIONS: In summary, HCMV-encoded US28 was detected in renal allografts from HCMV-positive donors independent of viremia and serostatus. Also, US28 facilitates HCMV spreading in VSMCs in vitro. Because the vasculature is affected in chronic renal transplant dysfunction, US28 may provide a potential target for therapeutic intervention.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/metabolism , Kidney Transplantation/adverse effects , Kidney/metabolism , Receptors, Chemokine/metabolism , Tissue Donors , Viral Proteins/metabolism , Adult , Aged , Allografts , Antibodies, Viral/blood , Biomarkers/blood , Biopsy , Cells, Cultured , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Humans , Immunoglobulin G/blood , Immunohistochemistry , Kidney/immunology , Kidney/surgery , Kidney/virology , Kidney Transplantation/methods , Male , Middle Aged , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/virology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/virology , Receptors, Chemokine/immunology , Retrospective Studies , Time Factors , Viral Proteins/immunology , Virulence , Young Adult
4.
Oncotarget ; 7(42): 67966-67985, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27602585

ABSTRACT

The human cytomegalovirus (HCMV) encoded chemokine receptor US28 promotes tumorigenesis through activation of various proliferative and angiogenic signaling pathways. Upon infection, US28 displays constitutive activity and signals in a G protein-dependent manner, hijacking the host's cellular machinery. In tumor cells, the hypoxia inducible factor-1α/pyruvate kinase M2 (HIF-1α/PKM2) axis plays an important role by supporting proliferation, angiogenesis and reprogramming of energy metabolism. In this study we show that US28 signaling results in activation of the HIF-1α/PKM2 feedforward loop in fibroblasts and glioblastoma cells. The constitutive activity of US28 increases HIF-1 protein stability through a Gαq-, CaMKII- and Akt/mTOR-dependent mechanism. Furthermore, we found that VEGF and lactate secretion are increased and HIF-1 target genes, glucose transporter type 1 (GLUT1) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), involved in glucose metabolism, are upregulated in US28 expressing cells. In addition, PKM2 is phosphorylated and found to be in a tumor-associated dimeric state upon US28 expression. Also in HCMV-infected cells HIF-1 activity is enhanced, which in part is US28-dependent. Finally, increased proliferation of cells expressing US28 is abolished upon inhibition of the HIF-1α/PKM2 cascade. These data highlight the importance of HIF-1α and PKM2 in US28-induced proliferation, angiogenesis and metabolic reprogramming.


Subject(s)
Carrier Proteins/metabolism , Glioblastoma/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Viral Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cytomegalovirus/physiology , Fibroblasts/metabolism , Fibroblasts/virology , Glioblastoma/genetics , Glioblastoma/virology , HEK293 Cells , Host-Pathogen Interactions , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Membrane Proteins/genetics , Mice , NIH 3T3 Cells , Receptors, Chemokine/genetics , Thyroid Hormones/genetics , Viral Proteins/genetics , Thyroid Hormone-Binding Proteins
5.
Methods Enzymol ; 570: 457-515, 2016.
Article in English | MEDLINE | ID: mdl-26921959

ABSTRACT

Chemokine receptors are involved in various pathologies such as inflammatory diseases, cancer, and HIV infection. Small molecule and antibody-based antagonists have been developed to inhibit chemokine-induced receptor activity. Currently two small molecule inhibitors targeting CXCR4 and CCR5 are on the market for stem cell mobilization and the treatment of HIV infection, respectively. Antibody fragments (e.g., nanobodies) targeting chemokine receptors are primarily orthosteric ligands, competing for the chemokine binding site. This is opposed by most small molecules, which act as allosteric modulators and bind to the receptor at a topographically distinct site as compared to chemokines. Allosteric modulators can be distinguished from orthosteric ligands by unique features, such as a saturable effect and probe dependency. For successful drug development, it is essential to determine pharmacological parameters (i.e., affinity, potency, and efficacy) and the mode of action of potential drugs during early stages of research in order to predict the biological effect of chemokine receptor targeting drugs in the clinic. This chapter explains how the pharmacological profile of chemokine receptor targeting ligands can be determined and quantified using binding and functional experiments.


Subject(s)
Chemokines/metabolism , Molecular Biology/methods , Molecular Targeted Therapy/methods , Receptors, Chemokine/metabolism , Allosteric Regulation , Animals , Binding, Competitive , Cell Line , Chemotaxis , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Iodine Radioisotopes , Ligands , Protein Binding , Receptors, Chemokine/agonists , Receptors, Chemokine/antagonists & inhibitors , Signal Transduction/drug effects , beta-Arrestins/metabolism
6.
Trends Pharmacol Sci ; 35(5): 247-55, 2014 May.
Article in English | MEDLINE | ID: mdl-24690241

ABSTRACT

G-protein-coupled receptors (GPCRs) represent a major therapeutic target class. A large proportion of marketed drugs exert their effect through modulation of GPCR function, and GPCRs have been successfully targeted with small molecules. Yet, the number of small new molecular entities targeting GPCRs that has been approved as therapeutics in the past decade has been limited. With new and improved immunization-related technologies and advances in GPCR purification and expression techniques, antibody-based targeting of GPCRs has gained attention. The serendipitous discovery of a unique class of heavy chain antibodies (hcAbs) in the sera of camelids may provide novel GPCR-directed therapies. Antigen-binding fragments of hcAbs, also referred to as nanobodies, combine the advantages of both small molecules (e.g., molecular cavity binding, low production costs) and monoclonal antibodies (e.g., high affinity and specificity). Nanobodies are gaining ground as therapeutics and are also starting to find application as diagnostics and as high-quality tools in GPCR research. Herein, we review recent advances in the use of nanobodies in GPCR research.


Subject(s)
Receptors, G-Protein-Coupled/immunology , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/immunology , Animals , Humans , Molecular Targeted Therapy , Receptors, G-Protein-Coupled/metabolism , Single-Domain Antibodies/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...