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










Publication year range
1.
J Cancer Res Clin Oncol ; 143(10): 2025-2038, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28667390

ABSTRACT

PURPOSE: Despite great progress in the diagnosis and treatment of localized prostate cancer (PCa), there remains a need for new diagnostic markers that can accurately distinguish indolent and aggressive variants. One promising approach is the antibody-based targeting of prostate stem cell antigen (PSCA), which is frequently overexpressed in PCa. Here, we show the construction of a molecular imaging probe comprising a humanized scFv fragment recognizing PSCA genetically fused to an engineered version of the human DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT), the SNAP-tag, enabling specific covalent coupling to various fluorophores for diagnosis of PCa. Furthermore, the recombinant immunotoxin (IT) PSCA(scFv)-ETA' comprising the PSCA(scFv) and a truncated version of Pseudomonas exotoxin A (PE, ETA') was generated. METHODS: We analyzed the specific binding and internalization behavior of the molecular imaging probe PSCA(scFv)-SNAP in vitro by flow cytometry and live cell imaging, compared to the corresponding IT PSCA(scFv)-ETA'. The cytotoxic activity of PSCA(scFv)-ETA' was tested using cell viability assays. Specific binding was confirmed on formalin-fixed paraffin-embedded tissue specimen of early and advanced PCa. RESULTS: Alexa Fluor® 647 labeling of PSCA(scFv)-SNAP confirmed selective binding to PSCA, leading to rapid internalization into the target cells. The recombinant IT PSCA(scFv)-ETA' showed selective binding leading to internalization and efficient elimination of target cells. CONCLUSIONS: Our data demonstrate, for the first time, the specific binding, internalization, and cytotoxicity of a scFv-based fusion protein targeting PSCA. Immunohistochemical staining confirmed the specific ex vivo binding to primary PCa material.


Subject(s)
ADP Ribose Transferases/administration & dosage , Antigens, Neoplasm/immunology , Bacterial Toxins/administration & dosage , Exotoxins/administration & dosage , Immunotoxins/administration & dosage , Neoplasm Proteins/immunology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Recombinant Fusion Proteins/administration & dosage , Virulence Factors/administration & dosage , ADP Ribose Transferases/immunology , Bacterial Toxins/immunology , Exotoxins/immunology , Flow Cytometry , GPI-Linked Proteins/immunology , HEK293 Cells , Humans , Immunologic Tests , Immunotherapy/methods , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Male , Molecular Targeted Therapy , Prostatic Neoplasms/immunology , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/immunology , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
2.
Int J Cancer ; 139(4): 916-27, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27037627

ABSTRACT

Chondroitin sulfate proteoglycan 4 (CSPG4) has been identified as a highly promising target antigen for immunotherapy of triple-negative breast cancer (TNBC). TNBC represents a highly aggressive heterogeneous group of tumors lacking expression of estrogen, progesterone and human epidermal growth factor receptor 2. TNBC is particularly prevalent among young premenopausal women. No suitable targeted therapies are currently available and therefore, novel agents for the targeted elimination of TNBC are urgently needed. Here, we present a novel cytolytic fusion protein (CFP), designated αCSPG4(scFv)-MAP, that consists of a high affinity CSPG4-specific single-chain antibody fragment (scFv) genetically fused to a functionally enhanced form of the human microtubule-associated protein (MAP) tau. Our data indicate that αCSPG4(scFv)-MAP efficiently targets CSPG4(+) TNBC-derived cell lines MDA-MB-231 and Hs 578T and potently inhibits their growth with IC50 values of ∼200 nM. Treatment with αCSPG(scFv)-MAP resulted in induction of the mitochondrial stress pathway by activation of caspase-9 as well as endonuclease G translocation to the nucleus, while induction of the caspase-3 apoptosis pathway was not detectable. Importantly, in vivo studies in mice bearing human breast cancer xenografts revealed efficient targeting to and accumulation of αCSPG4(scFv)-MAP at tumor sites resulting in prominent tumor regression. Taken together, this preclinical proof of concept study confirms the potential clinical value of αCSPG4(scFv)-MAP as a novel targeted approach for the elimination of CSPG4-positive TNBC.


Subject(s)
Antibodies, Monoclonal/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Membrane Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Triple Negative Breast Neoplasms/metabolism , tau Proteins/metabolism , Animals , Biomarkers , Biomarkers, Tumor , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chondroitin Sulfate Proteoglycans/genetics , Disease Models, Animal , Female , Gene Expression , Humans , Membrane Proteins/genetics , Mice , Molecular Targeted Therapy , Protein Binding , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tubulin/metabolism , Xenograft Model Antitumor Assays , tau Proteins/genetics
3.
Cancer Lett ; 372(2): 201-9, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-26806809

ABSTRACT

Triple-negative breast cancer (TNBC) is associated with poor prognosis and high prevalence among young premenopausal women. Unlike in other breast cancer subtypes, no targeted therapy is currently available. Overexpression of epithelial cell adhesion molecule (EpCAM) in 60% of TNBC tumors correlates with poorer prognosis and is associated with cancer stem cell phenotype. Thus, selective elimination of EpCAM(+) TNBC tumor cells is of clinical importance. Therefore, we constructed a fully human targeted cytolytic fusion protein, designated GbR201K-αEpCAM(scFv), in which an EpCAM-selective single-chain antibody fragment (scFv) is genetically fused to a granzyme B (Gb) mutant with reduced sensitivity to its natural inhibitor serpin B9. In vitro studies confirmed its specific binding, internalization and cytotoxicity toward a panel of EpCAM-expressing TNBC cells. Biodistribution kinetics and tumor-targeting efficacy using MDA-MB-468 cells in a human TNBC xenograft model in mice revealed selective accumulation of GbR201K-αEpCAM(scFv) in the tumors after i.v. injection. Moreover, treatment of tumor-bearing mice demonstrated a prominent inhibition of tumor growth of up to 50 % in this proof-of-concept study. Taken together, our results indicate that GbR201K-αEpCAM(scFv) is a promising novel targeted therapeutic for the treatment of TNBC.


Subject(s)
Apoptosis/drug effects , Cell Adhesion Molecules/antagonists & inhibitors , Granzymes/pharmacology , Immunotherapy/methods , Immunotoxins/pharmacology , Single-Chain Antibodies/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tumor Burden/drug effects , Animals , Antibody Specificity , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cell Adhesion Molecule , Female , Granzymes/genetics , Granzymes/pharmacokinetics , HEK293 Cells , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Mice, Inbred BALB C , Mice, Nude , Mutation , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution , Transfection , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Appl Microbiol Biotechnol ; 100(1): 263-77, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26428235

ABSTRACT

Autocrine activation of c-kit (KIT receptor tyrosine kinase) has been postulated to be a potent oncogenic driver in small cell lung cancer, neuroblastoma (NB), and poorly differentiated colorectal carcinoma (CRC). Although targeted therapy involving tyrosine kinase inhibitors (TKIs) such as imatinib mesylate is highly effective for gastrointestinal stromal tumor carrying V560G c-kit mutation, it does not show much potential for targeting wild-type KIT (WT-KIT). Our study demonstrates the role of stem cell factor (SCF)-based toxin conjugates for targeting WT-KIT-overexpressing malignancies such as NBs and CRCs. We constructed SCF-based recombinant bacterial toxins by genetically fusing mutated form of natural ligand SCF to receptor binding deficient forms of Diphtheria toxin (DT) or Pseudomonas exotoxin A (ETA') and evaluated their efficacy in vitro. Efficient targeting was achieved in all receptor-positive neuroblastoma (IMR-32 and SHSY5Y) and colon cancer cell lines (COLO 320DM, HCT 116, and DLD-1) but not in receptor-negative breast carcinoma cell line (MCF-7) thereby proving specificity. While dose- and time-dependent cytotoxicity was observed in both neuroblastoma cell lines, COLO 320DM and HCT 116 cells, only an anti-proliferative effect was observed in DLD-1 cells. We prove that these novel targeting agents have promising potential as KIT receptor tyrosine kinase targeting system.


Subject(s)
Antineoplastic Agents/metabolism , Bacterial Toxins/metabolism , Colorectal Neoplasms , Drug Delivery Systems/methods , Neuroblastoma , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Stem Cell Factor/metabolism , Bacterial Toxins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Stem Cell Factor/genetics
5.
Cancer Lett ; 365(2): 149-55, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25888452

ABSTRACT

The treatment of rhabdomyosarcoma (RMS) is challenging, and the prognosis remains especially poor for high-grade RMS with metastasis. The conventional treatment of RMS is based on multi-agent chemotherapy combined with resection and radiotherapy, which are often marked by low success rate. Alternative therapeutic options include the combination of standard treatments with immunotherapy. We generated a microtubule-associated protein (MAP)-based fully human cytolytic fusion protein (hCFP) targeting the fetal acetylcholine receptor, which is expressed on RMS cells. We were able to express and purify functional scFv35-MAP from Escherichia coli cells. Moreover, we found that scFv35-MAP is rapidly internalized by target cells after binding its receptor, and exhibits specific cytotoxicity toward FL-OH1 and RD cells in vitro. We also confirmed that scFv35-MAP induces apoptosis in FL-OH1 and RD cells. The in vivo potential of scFv35-MAP will need to be considered in further studies.


Subject(s)
Apoptosis/drug effects , Cholinergic Antagonists/pharmacology , Microtubule-Associated Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Rhabdomyosarcoma/drug therapy , Camptothecin/pharmacology , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Immunotherapy , Protein Binding , Receptors, Cholinergic/metabolism , Recombinant Fusion Proteins/genetics , U937 Cells
6.
J Cancer Res Clin Oncol ; 141(6): 1049-61, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25433506

ABSTRACT

PURPOSE: Rhabdomyosarcoma (RMS) is a rare and aggressive soft tissue sarcoma with limited treatment options and a high failure rate during standard therapy. New therapeutic strategies based on targeted immunotherapy are therefore much in demand. The epidermal growth factor receptor (EGFR) has all the characteristics of an ideal target. It is overexpressed in up to 80 % of embryonal RMS and up to 50 % of alveolar RMS tumors. We therefore tested the activity of the EGFR-specific recombinant immunotoxin (IT) 425(scFv)-ETA' against EGFR(+) RMS cells in vitro and ex vivo. METHODS: We tested the specific binding and internalization behavior of 425(scFv)-ETA' in RMS cell lines in vitro by flow cytometry, compared to the corresponding imaging probe 425(scFv)-SNAP monitored by live cell imaging. The cytotoxic activity of 425(scFv)-ETA' was tested using cell viability and apoptosis assays. Specific binding of the IT was confirmed on formalin-fixed paraffin-embedded tissue samples from two RMS patients. RESULTS: We confirmed the specific binding of 425(scFv)-ETA' to RMS cells in vitro and ex vivo. Both the IT and the corresponding imaging probe were rapidly internalized. The IT killed EGFR(+) RMS cells in a dose-dependent manner, while showing no effect against control cells. It showed specific apoptotic activity against one selected RMS cell line. CONCLUSIONS: This is the first study showing the promising therapeutic potential of a recombinant, EGFR-targeting, ETA'-based IT on RMS cells. We confirmed the selective killing with IC50 values of up to 50 pM, and immunohistochemical staining confirmed the specific ex vivo binding to primary RMS material.


Subject(s)
ErbB Receptors/metabolism , Immunotherapy/methods , Immunotoxins/pharmacology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , In Vitro Techniques , Microscopy, Confocal , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma, Alveolar/drug therapy , Rhabdomyosarcoma, Alveolar/pathology , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/pathology , Treatment Outcome , Up-Regulation
7.
Theranostics ; 4(10): 960-71, 2014.
Article in English | MEDLINE | ID: mdl-25157277

ABSTRACT

AIM: Fluorescence-mediated tomography (FMT) holds potential for accelerating diagnostic and theranostic drug development. However, for proper quantitative fluorescence reconstruction, knowledge on optical scattering and absorption, which are highly heterogeneous in different (mouse) tissues, is required. We here describe methods to assess these parameters using co-registered micro Computed Tomography (µCT) data and nonlinear whole-animal absorption reconstruction, and evaluate their importance for assessment of the biodistribution and target site accumulation of fluorophore-labeled drug delivery systems. METHODS: Besides phantoms with varying degrees of absorption, mice bearing A431 tumors were imaged 15 min and 48 h after i.v. injection of a fluorophore-labeled polymeric drug carrier (pHPMA-Dy750) using µCT-FMT. The outer shape of mice and a scattering map were derived using automated segmentation of the µCT data. Furthermore, a 3D absorption map was reconstructed from the trans-illumination data. We determined the absorption of five interactively segmented regions (heart, liver, kidney, muscle, tumor). Since blood is the main near-infrared absorber in vivo, the absorption was also estimated from the relative blood volume (rBV), determined by contrast-enhanced µCT. We compared the reconstructed absorption with the rBV-based values and analyzed the effect of using the absorption map on the fluorescence reconstruction. RESULTS: Phantom experiments demonstrated that absorption reconstruction is possible and necessary for quantitative fluorescence reconstruction. In vivo, the reconstructed absorption showed high values in strongly blood-perfused organs such as the heart, liver and kidney. The absorption values correlated strongly with the rBV-based absorption values, confirming the accuracy of the absorption reconstruction. Usage of homogenous absorption instead of the reconstructed absorption map resulted in reduced values in the heart, liver and kidney, by factors of 3.5, 2.1 and 1.4, respectively. For muscle and subcutaneous tumors, which have a much lower rBV and absorption, absorption reconstruction was less important. CONCLUSION: Quantitative whole-animal absorption reconstruction is possible and can be validated in vivo using the rBV. Usage of an absorption map is important when quantitatively assessing the biodistribution of fluorescently labeled drugs and drug delivery systems, to avoid a systematic underestimation of fluorescence in strongly absorbing organs, such as the heart, liver and kidney.


Subject(s)
Fluorescent Dyes/pharmacokinetics , X-Ray Microtomography/methods , Animals , Cell Line, Tumor , Fluorescent Dyes/chemistry , Humans , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasm Transplantation , Phantoms, Imaging , Spectrometry, Fluorescence , Tissue Distribution
8.
Cancer Lett ; 352(2): 228-35, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25016058

ABSTRACT

The treatment of rhabdomyosarcoma (RMS) remains challenging, with metastatic and alveolar RMS offering a particularly poor prognosis. Therefore, the identification and evaluation of novel antigens, which are suitable targets for immunotherapy, is one attractive possibility to improve the treatment of this disease. Here we show that chondroitin sulfate proteoglycan 4 (CSPG4) is expressed on RMS cell lines and RMS patient material. We evaluated the immunotoxin (IT) αMCSP-ETA', which specifically recognizes CSPG4 on the RMS cell lines RD, FL-OH1, TE-671 and Rh30. It is internalized rapidly, induces apoptosis and thus kills RMS cells selectively. We also demonstrate the specific binding of this IT to RMS primary tumor material from three different patients.


Subject(s)
ADP Ribose Transferases/pharmacology , Apoptosis/drug effects , Bacterial Toxins/pharmacology , Chondroitin Sulfate Proteoglycans/immunology , Exotoxins/pharmacology , Immunotoxins/pharmacology , Membrane Proteins/immunology , Rhabdomyosarcoma/pathology , Single-Chain Antibodies/pharmacology , Virulence Factors/pharmacology , ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Dose-Response Relationship, Drug , Exotoxins/metabolism , Humans , Immunotoxins/immunology , Immunotoxins/metabolism , Inhibitory Concentration 50 , Membrane Proteins/metabolism , Protein Binding , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/metabolism , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , Time Factors , Virulence Factors/metabolism , Pseudomonas aeruginosa Exotoxin A
9.
Curr Pharm Des ; 19(30): 5406-13, 2013.
Article in English | MEDLINE | ID: mdl-23431982

ABSTRACT

Over the past few years, the SNAP-tag technology has become a methodology with great potential in a variety of applications, e.g. the (specific) visualization of individual proteins and studies of protein interaction in living cells. Furthermore, the tag can be used for immunopurification and detection of recombinant proteins or site-specific coupling of recombinant proteins to surfaces. Next to the in vitro applications, it also enables detection of tagged proteins in vivo. This review gives an overview of the SNAP-tag technology in different fields of research and its potential for future developments.


Subject(s)
Molecular Imaging/methods , Proteins/chemistry , Proteins/metabolism , Animals , Gene Expression Regulation/physiology , Protein Binding , Proteins/genetics , Recombinant Proteins
10.
Curr Pharm Des ; 19(30): 5429-36, 2013.
Article in English | MEDLINE | ID: mdl-23431985

ABSTRACT

Although current cancer treatment strategies are highly aggressive, they are often not effective enough to destroy the collectivity of malignant cells. The residual tumor cells that survived the first-line treatment may continue to proliferate or even metastasize. Therefore, the development of novel more effective strategies to specifically eliminate also single cancer cells is urgently needed. In this respect, the development of antibody-based therapeutics, in particular example immunotoxins, has attracted broad interest. Since the internalization of immunotoxins is essential for their cytotoxic effectivity, it is of crucial importance to study their internalization behavior to assess the potential for their therapeutic use. In this study, we determined the internalization behavior of four different single-chain fragments variable (scFv) when binding to the corresponding target antigen as expressed on solid or non-solid tumor cell lines. The scFvs were recombinantly fused to the SNAP-tag, an engineered variant of the human repair enzyme O(6)-alkylguanine-DNA alkyltransferase that covalently reacts with benzylguanine derivatives. Since a large number of highly sensitive organic fluorescent dyes are already available or can easily be derivatized to react with the self-labeling SNAP-tag, this system provides versatile applications for imaging of intraand extracellular compartments of living cells. The fusion proteins were coupled to SNAP-surface(®) Alexa Fluor(®) 488 or SNAP-surface(®) Alexa Fluor(®) 647 and binding as well as internalization was monitored by flow cytometry and confocal microscopy, respectively. Depending on the respective target antigen, we could distinguish between slow and rapid internalization behavior. Moreover, we detected increased internalization rate for bivalent scFv constructs. Our approach allows for rapid and early stage evaluation of the internalization characteristics of new antibodies designated for further therapeutic development.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Molecular Imaging/methods , Neoplasms/metabolism , Animals , Antibodies , Cloning, Molecular , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Neoplasms/drug therapy , Protein Binding
11.
Cancer Immunol Immunother ; 61(10): 1617-26, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22350071

ABSTRACT

PURPOSE: Preclinical in vivo analyses of treatment responses are an important prerequisite to evaluate new therapeutics. Molecular in vivo imaging in the far red (FR)/near infra red (NIR) is a promising method, as it enables measurements at different time points in individual animals, thereby reducing the number of animals required, while increasing statistical significance. Here, we show the establishment of a method to monitor response to treatment using fluorescent cells, expressing the epidermal growth factor receptor (EGFR), a target already used in therapy. METHODS: We transfected A-431 tumour cells with the far red-emitting protein Katushka (Kat2), resulting in strong fluorescence allowing for the monitoring of tumour growth when implanted in BALB/c nu/nu mice with a CRi Maestro in vivo imager. We targeted A-431 cells with a previously reported immunotoxin (IT), consisting of the anti-EGFR antibody single-chain variable fragment (scFv) 425, fused to Pseudomonas aeruginosa Exotoxin A' (ETA'). In addition, EGFR expression was verified using the 425(scFv) conjugated to a NIR dye BG-747 through a SNAP-tag linker. RESULTS: The results show the feasibility to evaluate response to treatment in vivo by FR imaging, while at the same location detecting EGFR expression. Treatment with 425(scFv)-ETA' resulted in decelerated tumour growth, while not affecting the overall health of the animals. This is in contrast to treatment with Doxorubicin, which, although decreasing the tumour size, resulted in poor health. CONCLUSIONS: We developed a novel method to non-invasively determine treatment responses by in vivo imaging of multiple parameters which showed the efficacy of 425(scFv)-ETA'.


Subject(s)
ADP Ribose Transferases/therapeutic use , Antineoplastic Agents/therapeutic use , Bacterial Toxins/therapeutic use , Carcinoma/drug therapy , Exotoxins/therapeutic use , Immunotoxins/therapeutic use , Luminescent Proteins/metabolism , Molecular Imaging/methods , Skin Neoplasms/drug therapy , Spectroscopy, Near-Infrared/methods , Virulence Factors/therapeutic use , Animals , Carcinoma/diagnosis , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/immunology , Female , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Imaging/instrumentation , Skin Neoplasms/diagnosis , Spectroscopy, Near-Infrared/instrumentation , Transfection , Xenograft Model Antitumor Assays , Red Fluorescent Protein , Pseudomonas aeruginosa Exotoxin A
12.
Protein Expr Purif ; 58(2): 325-31, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18261921

ABSTRACT

The Ca2+-dependent binding of annexin A5 to phosphatidylserine on cell surfaces is a reliable marker for apoptosis that is widely used in flow cytometry based apoptosis assays. In this approach, annexin A5 must be coupled to a fluorescent dye, but standard dyes such as fluorescein are photolabile, and the heterogeneous chemical linkage partially inhibits binding to phosphatidylserine. Recombinant fusions comprising annexin A5 and fluorescent proteins are available for prokaryotic expression, but can be purified only at low concentrations due to their low solubility in the cytoplasm. Here we describe a eukaryotic expression system for the secretion of functional recombinant annexin A5, with and without fluorescent protein fusions, in different formats. Metal affinity purification yielded up to 18 microg of histidine-tagged annexin A5 fusions per ml processed cell culture supernatants. Furthermore the supernatant itself was sufficient for direct use in apoptosis assays. The availability of such fusion proteins offers new and more economical opportunities for the development and application of this widely utilized apoptosis assay.


Subject(s)
Annexin A5/biosynthesis , Green Fluorescent Proteins/chemistry , Annexin A5/metabolism , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Humans , Kidney , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...