Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Carcinogenesis ; 31(11): 1913-21, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20837599

ABSTRACT

The epithelial cell adhesion molecule (EpCAM) is a membrane glycoprotein that is highly expressed on most carcinomas and therefore of potential use as a diagnostic and prognostic marker for a variety of carcinomas. Interestingly, EpCAM is explored as target in antibody-based therapies. Recently, EpCAM has been identified as an additional marker of cancer-initiating cells. In this review, we describe the controversial biological role of EpCAM with the focus on carcinogenesis: as an adhesion molecule, EpCAM mediates homophilic adhesion interactions, which in turn might prevent metastasis. On the other hand, EpCAM abrogates E-cadherin mediated cell-cell adhesion thereby promoting metastasis. Also, upon cleavage of EpCAM, the intracellular domain functions as a part of a transcriptional complex inducing c-myc and cyclin A and E. In line with these seemingly controversial roles, EpCAM overexpression has been associated with both decreased and increased survival of patients. Similarly, either induction or downregulation of EpCAM expression lowers the oncogenic potential depending on the cell type. As epigenetic dysregulation underlies aberrant EpCAM expression, we propose epigenetic editing as a novel approach to investigate the biological role of EpCAM, expanding the options for EpCAM as a therapeutic target in cancer.


Subject(s)
Antigens, Neoplasm/physiology , Cell Adhesion Molecules/physiology , Neoplasms/metabolism , Cell Transformation, Neoplastic , Epigenesis, Genetic , Epithelial Cell Adhesion Molecule , Humans
2.
Exp Cell Res ; 315(15): 2487-95, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19540229

ABSTRACT

TRAIL induced apoptosis of tumor cells is currently entering phase II clinical settings, despite the fact that not all tumor types are sensitive to TRAIL. TRAIL resistance in ovarian carcinomas can be caused by a blockade upstream of the caspase 3 signaling cascade. We explored the ability of restriction endonucleases to directly digest DNA in vivo, thereby circumventing the caspase cascade. For this purpose, we delivered enzymatically active endonucleases via the cationic amphiphilic lipid SAINT-18((R)):DOPE to both TRAIL-sensitive and insensitive ovarian carcinoma cells (OVCAR and SKOV-3, respectively). Functional nuclear localization after delivery of various endonucleases (BfiI, PvuII and NucA) was indicated by confocal microscopy and genomic cleavage analysis. For PvuII, analysis of mitochondrial damage demonstrated extensive apoptosis both in SKOV-3 and OVCAR. This study clearly demonstrates that cellular delivery of restriction endonucleases holds promise to serve as a novel therapeutic tool for the treatment of resistant ovarian carcinomas.


Subject(s)
Apoptosis/physiology , Endonucleases/metabolism , Ovarian Neoplasms , TNF-Related Apoptosis-Inducing Ligand/metabolism , Amino Acid Chloromethyl Ketones/metabolism , Caspase 3/metabolism , Cell Line, Tumor , DNA Fragmentation , Enzyme Inhibitors/metabolism , Female , Fibroblasts/cytology , Fibroblasts/physiology , Histones/metabolism , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphatidylethanolamines/metabolism , Pyridinium Compounds/metabolism , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/genetics
3.
Front Biosci ; 13: 5050-5, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508569

ABSTRACT

Embryonic development is one of the most complex biological phenomena that involves the appropriate expression and synchronized interactions of a plethora of proteins, including cell adhesion molecules (CAMs). Many members of the diverse family of CAMs have been shown to be critically involved in the correct execution of embryonic development. The Epithelial Cell Adhesion Molecule (EpCAM) is an atypical cell adhesion molecule originally identified as a marker for carcinoma. However, recent insights have revealed that EpCAM participates in not only cell adhesion, but also in proliferation, migration and differentiation of cells. All of these processes are known to be fundamental for morphogenesis. Here, we review the current literature that establishes EpCAM as a protein involved in morphogenesis, starting from the earliest stages of embryogenesis and ending in organogenesis. In addition, we provide directions for further elucidation of the role of EpCAM in embryogenesis.


Subject(s)
Antigens, Neoplasm/physiology , Cell Adhesion Molecules/physiology , Embryonic Development , Morphogenesis/physiology , Organogenesis/physiology , Animals , Epithelial Cell Adhesion Molecule , Humans , Kidney/embryology , Lung/embryology , Pancreas/embryology
4.
J Mol Med (Berl) ; 86(8): 909-24, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18504532

ABSTRACT

Previously, we have shown that epidermal growth factor receptor (EGFR)-selective delivery of soluble tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL), by genetic fusion to antibody fragment scFv425, enhances the tumor-selective pro-apoptotic activity of sTRAIL. Insight into the respective contribution of the agonistic receptors TRAIL-R1 and TRAIL-R2 to TRAIL-induced apoptosis may provide a rational approach to further optimize TRAIL-based therapy. Recently, this issue has been investigated using sTRAIL mutants designed to selectively bind to either receptor. However, the relative contribution of the respective TRAIL receptors, in particular TRAIL-R1, in TRAIL signaling is still unresolved. Here, we fused scFv425 to designed sTRAIL mutant sTRAILmR1-5, reported to selectively activate TRAIL-R1, and investigated the therapeutic apoptotic activity of this novel fusion protein. EGFR-specific binding of scFv425:sTRAILmR1-5 potently induced apoptosis, which was superior to the apoptotic activity of scFv425:sTRAIL-wt and a nontargeted MOCK-scFv:sTRAILmR1-5. During cotreatment with cisplatin or the histone deacetylase inhibitor valproic acid, scFv425:sTRAILmR1-5 retained its superior pro-apoptotic activity compared to scFv425:sTRAIL-wt. However, in catching-type Enzyme-Linked ImmunoSorbent Assays with TRAIL-R1:Fc and TRAIL-R2:Fc, scFv425:sTRAILmR1-5 was found to not only bind to TRAIL-R1 but also to TRAIL-R2. Binding to TRAIL-R2 also had functional consequences because the apoptotic activity of scFv425:sTRAILmR1-5 was strongly inhibited by a TRAIL-R2 blocking monoclonal antibody. Moreover, scFv425:sTRAILmR1-5 retained apoptotic activity upon selective knockdown of TRAIL-R1 using small inhibitory RNA. Collectively, these data indicate that both agonistic TRAIL receptors are functionally involved in TRAIL signaling by scFv425:sTRAILmR1-5 in solid tumor cells. Moreover, the superior target cell-restricted apoptotic activity of scFv425:sTRAILmR1-5 indicates its therapeutic potential for EGFR-positive solid tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis , ErbB Receptors/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Antibodies, Monoclonal/metabolism , Binding Sites , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Gene Transfer Techniques , Humans , Jurkat Cells , Ligands , Mutation , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/therapeutic use
5.
Int J Cancer ; 123(2): 484-489, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18398839

ABSTRACT

The epithelial cell adhesion molecule (EpCAM) is expressed at high levels on the surface of most carcinoma cells. SiRNA silencing of EpCAM expression leads to reduced metastatic potential of tumor cells demonstrating its importance in oncogenesis and tumor progression. However, siRNA therapy requires either sequential delivery or integration into the host cell genome. Hence we set out to explore a more definite form to influence EpCAM gene expression. The mechanisms underlying the transcriptional activation of the EpCAM gene, both in normal epithelial tissue as well as in carcinogenesis, are poorly understood. We show that DNA methylation plays a crucial role in EpCAM expression, and moreover, active silencing of endogenous EpCAM via methylation of the EpCAM promoter results in a persistent downregulation of EpCAM expression. In a panel of carcinoma derived cell lines, bisulfite analyses showed a correlation between the methylation status of the EpCAM promoter and EpCAM expression. Treatment of EpCAM-negative cell lines with a demethylating agent induced EpCAM expression, both on mRNA and protein level, and caused upregulation of EpCAM expression in an EpCAM-positive cell line. After delivery of the DNA methyltransferase M.SssI into EpCAM-positive ovarian carcinoma cells, methylation of the EpCAM promoter resulted in silencing of EpCAM expression. SiRNA-mediated silencing remained for 4 days, after which EpCAM re-expression increased in time, while M.SssI-mediated downregulation of EpCAM maintained through successive cell divisions as the repression persisted for at least 17 days. This is the first study showing that active DNA methylation leads to sustained silencing of endogenous EpCAM expression.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma/metabolism , Cell Adhesion Molecules/metabolism , Cell Nucleus/metabolism , DNA Methylation , Antigens, Neoplasm/genetics , Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Carcinoma/drug therapy , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , DNA Methylation/drug effects , DNA-Cytosine Methylases/metabolism , Down-Regulation , Epithelial Cell Adhesion Molecule , Female , Gene Expression Regulation, Neoplastic , Gene Silencing/drug effects , Humans , Ovarian Neoplasms/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic , Up-Regulation/drug effects
6.
Transgenic Res ; 17(2): 229-38, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17940847

ABSTRACT

The Epithelial Cell Adhesion Molecule (EpCAM) is expressed virtually on normal epithelia in vertebrates. Among different species, the amino acid sequence of EpCAM is highly homologous, indicating that EpCAM is an evolutionary conserved protein. However, differences in the expression pattern of EpCAM homologues have been reported. We hypothesized that differences in expression pattern might be related to the promoter organization of the respective EpCAM homologues. Therefore, we here compared the promoter region of the mouse and human EpCAM homologues. In addition, we compared the expression pattern of the human and murine EpCAM homologues in the hEpCAM transgenic mouse. In silico analysis of EpCAM homologues revealed that the amino acid sequence as well as the domain structure is highly conserved throughout different vertebrates. In silico analysis of the promoter region identified that the human and mouse EpCAM promoters share a low homology. In agreement with this low homology, the murine and human EpCAM promoter contains only a few common transcription factor binding sites. Nevertheless, immunohistochemcial analysis of the expression of human and murine EpCAM in lung, colon, and kidney of the hEpCAM transgenic mouse identified that expression of both homologues is restricted to epithelial cells in these organs. Moreover, in lung and colon the human and murine homologues of EpCAM were co-expressed. In contrast, the EpCAM homologues were only sporadically co-expressed in renal epithelia, although they were distributed similarly along the nephronic segments. Together, these findings indicate an overall conserved regulatory mechanism that ensures epithelial expression of EpCAM homologues, despite the low promoter homology. Furthermore, the fact that murine epithelia express the human homologue of EpCAM indicates that the mouse has transcription factors required for human EpCAM expression.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Kidney/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Colon/cytology , Colon/metabolism , Computational Biology , Epithelial Cell Adhesion Molecule , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Kidney/cytology , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Transcription Factors/metabolism , Transcription, Genetic
7.
Nephron Exp Nephrol ; 107(4): e119-31, 2007.
Article in English | MEDLINE | ID: mdl-18025791

ABSTRACT

BACKGROUND: The epithelial cell adhesion molecule (EpCAM) is expressed by most epithelia and is involved in processes fundamental for morphogenesis, including cell-cell adhesion, proliferation, differentiation, and migration. Previously, a role for EpCAM in pancreatic morphogenesis was confirmed in vitro. Furthermore, changes in the EpCAM expression pattern were found in developing lung and thymus and in the regenerating liver. Therefore, EpCAM was proposed to be a morphoregulatory molecule. METHODS: Using immunohistochemistry, the expression pattern of human and murine homologues of EpCAM was characterized in adult and embryonic kidneys from humans and human-EpCAM (hEpCAM)-transgenic mice. RESULTS: EpCAM expression was found in the ureteric bud throughout nephrogenesis. EpCAM was not expressed in the metanephric mesenchyme. In comma- and S-shaped bodies, both metanephric mesenchyme derived structures, EpCAM expression appeared by E13.5. In adult kidneys, most epithelia expressed varying levels of EpCAM, as confirmed by double staining for human EpCAM and segment-specific nephron markers. Podocytes were EpCAM negative. At the cellular level, the EpCAM expression shifted from apical in embryonic to basolateral in adult kidneys. CONCLUSIONS: The spatiotemporal expression pattern of EpCAM changes during nephrogenesis. In the adult kidney, the expression varies markedly along the nephron. These data provide a basis for further studies on EpCAM in developing and adult kidneys.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Kidney , Adult , Age Factors , Animals , Animals, Newborn , Antibodies, Monoclonal , Antigens, Neoplasm/immunology , CHO Cells , Cell Adhesion Molecules/immunology , Cell Polarity , Cricetinae , Cricetulus , Epithelial Cell Adhesion Molecule , Epithelial Cells/cytology , Female , Humans , Immunohistochemistry , Kidney/embryology , Kidney/growth & development , Kidney/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Staining and Labeling
8.
J Control Release ; 123(3): 228-38, 2007 Nov 20.
Article in English | MEDLINE | ID: mdl-17884225

ABSTRACT

Cationic liposomal compounds are widely used to introduce DNA and siRNA into viable cells, but none of these compounds are also capable of introducing proteins. Here we describe the use of a cationic amphiphilic lipid SAINT-2:DOPE for the efficient delivery of proteins into cells (profection). Labeling studies demonstrated equal delivery efficiency for protein as for DNA and siRNA. Moreover, proteins complexed with Saint-2:DOPE were successfully delivered, irrespective of the presence of serum, and the profection efficiency was not influenced by the size or the charge of the protein:cationic liposomal complex. Using beta-galactosidase as a reporter protein, enzymatic activity was detected in up to 98% of the adherent cells, up to 83% of the suspension cells and up to 70% of the primary cells after profection. A delivered antibody was detected in the cytoplasm for up to 7 days after profection. Delivery of the methyltransferase M.SssI resulted in DNA methylation, leading to a decrease in E-cadherin expression. The lipid-mediated multipurpose transport system reported here can introduce proteins into the cell with an equal delivery efficiency as for nucleotides. Delivery is irrespective of the presence of serum, and the protein can exert its function both in the cytoplasm and in the nucleus. Furthermore, DNA methylation by M.SssI delivery as a novel tool for gene silencing has potential applications in basic research and therapy.


Subject(s)
Cell Nucleus/metabolism , Drug Carriers , Phosphatidylethanolamines/chemistry , Proteins/metabolism , Pyridinium Compounds/chemistry , Serum/metabolism , Active Transport, Cell Nucleus , Animals , Antibodies/metabolism , COS Cells , Cadherins/genetics , Cadherins/metabolism , Cations , Cell Nucleus/enzymology , Chemistry, Pharmaceutical , Chlorocebus aethiops , DNA/metabolism , DNA Methylation , DNA-Cytosine Methylases/metabolism , Drug Compounding , Gene Silencing , Humans , Jurkat Cells , Molecular Structure , Particle Size , Protein Conformation , Proteins/chemistry , Proteins/genetics , RNA, Small Interfering/metabolism , Time Factors , Transfection , beta-Galactosidase/metabolism
9.
J Immunol ; 179(2): 1362-8, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617629

ABSTRACT

The human pancarcinoma-associated epithelial cell adhesion molecule (EpCAM) (EGP-2, CO17-1A) is a well-known target for carcinoma-directed immunotherapy. Mouse-derived mAbs directed to EpCAM have been used to treat colon carcinoma patients showing well-tolerable toxic side effects but limited antitumor effects. Humanized or fully human anti-EpCAM mAbs may induce stronger antitumor activity, but proved to produce severe pancreatitis upon use in patients. To evaluate treatment-associated effects before a clinical trial, we have generated a transgenic mouse tumor model that expresses human EpCAM similar to carcinoma patients. In this study, we use this model to study the in vivo behavior of two humanized and one mouse-derived anti-EpCAM mAb, i.e., MOC31-hFc, UBS54, and MOC31. The pharmacokinetics and tissue distribution of the fully human mAb UBS54 and the mouse-derived MOC31 were largely the same after injection in tumor-bearing transgenic mice, whereas the molecularly engineered, humanized MOC31-hFc behaved differently. Injection of UBS54 and MOC31 resulted in significant, dose-dependent uptake of mAb in EpCAM-expressing normal and tumor tissues, accompanied by a drop in serum level, whereas injection of MOC31-hFc resulted in uptake in tumor tissue, limited uptake by normal tissues, and slow blood clearance. It is concluded that the EpCAM-transgenic mouse model provides valuable insights into the potential behavior of humanized anti-EpCAM mAbs in patients. mAbs sharing the same epitope and isotype but constructed differently were shown to behave differently in the model, indicating that the design of mAbs is important for eventual success in in vivo application.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Immunotherapy/methods , Neoplasms, Experimental/immunology , Animals , Antibodies, Monoclonal/immunology , Disease Models, Animal , Epithelial Cell Adhesion Molecule , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Tissue Distribution
10.
Am J Pathol ; 171(2): 386-95, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17600130

ABSTRACT

The epithelial cell adhesion molecule (EpCAM, CD326) is a glycoprotein of approximately 40 kd that was originally identified as a marker for carcinoma, attributable to its high expression on rapidly proliferating tumors of epithelial origin. Normal epithelia express EpCAM at a variable but generally lower level than carcinoma cells. In early studies, EpCAM was proposed to be a cell-cell adhesion molecule. However, recent insights revealed a more versatile role for EpCAM that is not limited only to cell adhesion but includes diverse processes such as signaling, cell migration, proliferation, and differentiation. Cell surface expression of EpCAM may actually prevent cell-cell adhesion. Here, we provide a comprehensive review of the current knowledge on EpCAM biology in relation to other cell adhesion molecules. We discuss the implications of the newly identified functions of EpCAM in view of its prognostic relevance in carcinoma, inflammatory pathophysiology, and tissue development and regeneration as well as its role in normal epithelial homeostasis.


Subject(s)
Antigens, Neoplasm/physiology , Cell Adhesion Molecules/physiology , Neoplasms/pathology , Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Carcinoma/metabolism , Carcinoma/pathology , Cell Adhesion Molecules/analysis , Cell Cycle , Cell Proliferation , Epithelial Cell Adhesion Molecule , Humans , Models, Biological , Neoplasms/metabolism , Signal Transduction
11.
J Immunol ; 178(4): 2287-95, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277134

ABSTRACT

Ab binding to CD20 has been shown to induce apoptosis in B cells. In this study, we demonstrate that rituximab sensitizes lymphoma B cells to Fas-induced apoptosis in a caspase-8-dependent manner. To elucidate the mechanism by which Rituximab affects Fas-mediated cell death, we investigated rituximab-induced signaling and apoptosis pathways. Rituximab-induced apoptosis involved the death receptor pathway and proceeded in a caspase-8-dependent manner. Ectopic overexpression of FLIP (the physiological inhibitor of the death receptor pathway) or application of zIETD-fmk (specific inhibitor of caspase-8, the initiator-caspase of the death receptor pathway) both specifically reduced rituximab-induced apoptosis in Ramos B cells. Blocking the death receptor ligands Fas ligand or TRAIL, using neutralizing Abs, did not inhibit apoptosis, implying that a direct death receptor/ligand interaction is not involved in CD20-mediated cell death. Instead, we hypothesized that rituximab-induced apoptosis involves membrane clustering of Fas molecules that leads to formation of the death-inducing signaling complex (DISC) and downstream activation of the death receptor pathway. Indeed, Fas coimmune precipitation experiments showed that, upon CD20-cross-linking, Fas-associated death domain protein (FADD) and caspase-8 were recruited into the DISC. Additionally, rituximab induced CD20 and Fas translocation to raft-like domains on the cell surface. Further analysis revealed that, upon stimulation with rituximab, Fas, caspase-8, and FADD were found in sucrose-gradient raft fractions together with CD20. In conclusion, in this study, we present evidence for the involvement of the death receptor pathway in rituximab-induced apoptosis of Ramos B cells with concomitant sensitization of these cells to Fas-mediated apoptosis via Fas multimerization and recruitment of caspase-8 and FADD to the DISC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Burkitt Lymphoma/immunology , Fas Ligand Protein/immunology , Receptor Aggregation/drug effects , fas Receptor/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20/immunology , Antineoplastic Agents/therapeutic use , Apoptosis/immunology , Burkitt Lymphoma/drug therapy , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , Caspase 8/immunology , Cell Line, Tumor , Fas-Associated Death Domain Protein/immunology , Humans , Membrane Microdomains/immunology , Protein Transport/drug effects , Protein Transport/immunology , Receptor Aggregation/immunology , Rituximab
12.
Cell Transplant ; 15(5): 381-7, 2006.
Article in English | MEDLINE | ID: mdl-16970280

ABSTRACT

Xenotransplantation of porcine fetal ventral mesencephalic (pfVM) cells to overcome the dopamine shortage in the striatum of patients with Parkinson's disease seems a viable alternative to allotransplantion of human fetal donor tissue, especially because the latter is complicated by both practical and ethical issues. There is, however, little known about the xenospecific immune responses involved in such an intracerebral xenotransplantation. The aim of our study was to investigate whether (1) naive human peripheral blood mononuclear cells (PMBC) display cytotoxicity against pfVM cells of E28 pig fetuses, and (2) priming of human PBMC by xenogeneic antigen presenting cells (APC) modulates pfVM-directed cellular cytotoxicity. For this purpose fresh PMBC from nine individual donors were primed by incubation with either irradiated pfVM cells or porcine spleen cells (PSC) as APC in the presence of IL-2 for 1 week before assessing cytotoxicity in a 51Cr release assay. Also, direct NK reactivity and antibody-dependent cellular cytotoxicity (ADCC) of fresh PMBC against pfVM cells was assessed. No direct cytotoxicity of naive cells (either NK reactivity or ADCC) against pfVM cells could be determined. Only PMBC primed with PSC were capable of lysing pfVM cells. PBMC primed with pfVM cells did not show cytolytic capacity towards pfVM. Interestingly, large differences in xenospecific T-cell responses exist between individual donor PBMC. Thus, human T cells are capable of killing pfVM cells in a xenoreactive response, but only after priming by donor APC. The large interindividual differences between human donors in their xenoreactive response may influence patient selection for xenotransplantation and chances of graft survival for individual patients.


Subject(s)
Fetal Tissue Transplantation/immunology , Mesencephalon/immunology , T-Lymphocytes/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/physiology , Antigen-Presenting Cells/metabolism , Cells, Cultured , Cytotoxicity Tests, Immunologic/methods , Fetal Tissue Transplantation/methods , Humans , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mesencephalon/cytology , Swine , T-Lymphocytes/metabolism , Transplantation, Heterologous
13.
Trends Mol Med ; 12(8): 382-93, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16798087

ABSTRACT

Important breakthroughs in cancer therapy include clinical application of antibodies, such as Rituximab, and small inhibitory molecules, such as Iressa and Velcade. In addition, recent reports have indicated the therapeutic potential of physiological pro-apoptotic proteins such as TRAIL and galectin-1. Although unrelated at first glance, each strategy relies on the deliberate and selective induction of apoptosis in malignant cells. Importantly, therapy-resistance in cancer is frequently associated with de-regulation in the mechanisms that control apoptosis. However, cancer cells are often reliant on these molecular aberrations for survival. Therefore, selective induction of apoptosis in cancer cells but not normal cells seems feasible. Here, we review recent progress and prospects of selected novel anti-cancer approaches that specifically target and sensitize cancer cells to apoptosis.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Neoplasms/drug therapy , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Galectins/pharmacology , Galectins/therapeutic use , Gefitinib , Humans , Models, Biological , Neoplasms/physiopathology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Rituximab , Signal Transduction/drug effects , Signal Transduction/physiology
14.
J Infect Dis ; 193(11): 1552-6, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16652284

ABSTRACT

Targeting viral proteins early during infection may limit exacerbation of human cytomegalovirus infection. The viral chemokine-receptor homologue US28 interferes with leukocyte trafficking and, possibly, viral replication. Because US28 molecules are abundant on the surface of infected cells, this homologue is a potential target for antiviral therapy. To assess the relationship between US28 and disease activity, we measured, by quantitative reverse-transcription polymerase chain reaction, the levels of US28 and immediate-early (IE) 1 gene transcripts in the blood of lung-transplant recipients. We found that, during primary and secondary infection, the IE1 and US28 genes have early transcription kinetics and are expressed at similar levels. This may render US28 an attractive target for antiviral therapy.


Subject(s)
Cytomegalovirus Infections/virology , Gene Expression , Lung Transplantation , Receptors, Chemokine/genetics , Viral Proteins/genetics , Adult , Antigens, Viral/blood , Humans , Immediate-Early Proteins/genetics , Kinetics , Middle Aged , Phosphoproteins/blood , RNA, Messenger/blood , RNA, Viral/blood , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Viral Matrix Proteins/blood
15.
Transfusion ; 46(4): 554-61, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16584431

ABSTRACT

BACKGROUND: Human mitochondrial DNA (mtDNA) polymorphisms can be used to detect allogeneic transfused platelets. To increase the number of informative polymorphisms we investigated three hypervariable regions (HVR1, HVR2, and HVR3) within the displacement loop (D-loop) region of the mtDNA. STUDY DESIGN AND METHODS: mtDNA was obtained from 119 unrelated blood donors. Forward and reverse primers were designed and conditions optimized to amplify and sequence the template mtDNA by dye terminator cycle sequencing. RESULTS: We established a sequencing protocol for all three HVRs of the mtDNA. Polymorphic sites were found in all three regions: 66 in HVR1, 44 in HVR2, and 18 in HVR3. Combining the sequence information of HVR1, -2, and -3 resulted in 105 different genotypes of which 95 were unique. We were able to discriminate between two randomly chosen individuals with a random match probability of 1.2 percent. CONCLUSION: The D-loop region of mtDNA contains a wealth of informative molecular markers for chimerism and survival studies after transfusions of cellular blood components.


Subject(s)
Complementarity Determining Regions/genetics , DNA, Mitochondrial/genetics , Genetic Variation , Platelet Transfusion , Base Sequence , DNA Primers , DNA, Mitochondrial/blood , DNA, Mitochondrial/isolation & purification , Humans , Probability , Transplantation, Homologous/physiology
16.
Virus Res ; 118(1-2): 196-200, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16448715

ABSTRACT

The activation of the major immediate-early promoter (MIEP) is a key event in the cytomegalovirus replication cycle and is dependent on cellular transcription factors which are partially activated by viral proteins. Expression of the viral chemokine receptor homolog US28 results in constitutive activation of pro-inflammatory transcription factors that may be involved in the activation of the major immediate-early promoter/enhancer. Using reporter gene assays in human embryonic kidney cells, we found that US28 signaling was responsible for increased major immediate-early promoter/enhancer activity which was independent of beta-chemokine binding. Inhibition of nuclear factor-kappaB (NF-kappaB) only partially blocked the effect of US28, whereas treatment with a specific p38 mitogen activated kinase (MAPK) inhibitor fully abrogated the US28-induced enhancement of promoter activity. Our results suggest that during human cytomegalovirus (HCMV) infection, US28 in epithelial cells transactivates the major immediate-early promoter/enhancer via the activation of p38 MAPK and downstream signaling that partially involves NF-kappaB.


Subject(s)
Cytomegalovirus/physiology , Enhancer Elements, Genetic , Gene Expression Regulation, Viral , Genes, Immediate-Early , Promoter Regions, Genetic , Receptors, Chemokine/physiology , Viral Proteins/physiology , Artificial Gene Fusion , Cell Line , Cytomegalovirus/genetics , Genes, Reporter , Humans , Luciferases/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Receptors, Chemokine/genetics , Signal Transduction , Viral Proteins/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Blood ; 107(7): 2863-70, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16332967

ABSTRACT

Agonistic anti-Fas antibodies and multimeric recombinant Fas ligand (FasL) preparations show high tumoricidal activity against leukemic cells, but are unsuitable for clinical application due to unacceptable systemic toxicity. Consequently, new antileukemia strategies based on Fas activation have to meet the criterion of strictly localized action at the tumor-cell surface. Recent insight into the FasL/Fas system has revealed that soluble homotrimeric FasL (sFasL) is in fact nontoxic to normal cells, but also lacks tumoricidal activity. We report on a novel fusion protein, designated scFvCD7:sFasL, that is designed to have leukemia-restricted activity. ScFvCD7:sFasL consists of sFasL genetically linked to a high-affinity single-chain fragment of variable regions (scFv) antibody fragment specific for the T-cell leukemia-associated antigen CD7. Soluble homotrimeric scFvCD7:sFasL is inactive and acquires tumoricidal activity only after specific binding to tumor cell-surface-expressed CD7. Treatment of T-cell acute lymphoblastic leukemia (T-ALL) cell lines and patient-derived T-ALL, peripheral T-cell lymphoma (PTCL), and CD7-positive acute myeloid leukemia (AML) cells with homotrimeric scFvCD7:sFasL revealed potent CD7-restricted induction of apoptosis that was augmented by conventional drugs, farnesyl transferase inhibitor L-744832, and the proteasome inhibitor bortezomib (Velcade; Millenium, Cambridge, MA). Importantly, identical treatment did not affect normal human peripheral-blood lymphocytes (PBLs) and endothelial cells, with only moderate apoptosis in interleukin-2 (IL-2)/CD3-activated T cells. CD7-restricted activation of Fas in T-cell leukemic cells by scFvCD7:sFasL revitalizes interest in the applicability of Fas signaling in leukemia therapy.


Subject(s)
Antigens, CD7/immunology , Apoptosis/immunology , Leukemia-Lymphoma, Adult T-Cell/therapy , Membrane Glycoproteins/immunology , Tumor Necrosis Factors/immunology , Antigens, CD7/genetics , Antigens, CD7/therapeutic use , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bystander Effect , Cell Line, Tumor , DNA Primers , Fas Ligand Protein , Humans , Jurkat Cells , Leukemia-Lymphoma, Adult T-Cell/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/therapeutic use , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/therapeutic use
18.
Cancer Res ; 65(8): 3380-8, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15833872

ABSTRACT

Current treatment of human T-cell leukemia and lymphoma is predominantly limited to conventional cytotoxic therapy and is associated with limited therapeutic response and significant morbidity. Therefore, more potent and leukemia-specific therapies with favorable toxicity profiles are urgently needed. Here, we report on the construction of a novel therapeutic fusion protein, scFvCD7:sTRAIL, designed to induce target antigen-restricted apoptosis in human T-cell tumors. ScFvCD7:sTRAIL consists of the death-inducing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) genetically linked to an scFv antibody fragment specific for the T-cell surface antigen CD7. Treatment with scFvCD7:sTRAIL induced potent CD7-restricted apoptosis in a series of malignant T-cell lines, whereas normal resting leukocytes, activated T cells, and vascular endothelial cells (human umbilical vein endothelial cells) showed no detectable apoptosis. The apoptosis-inducing activity of scFvCD7:sTRAIL was stronger than that of the immunotoxin scFvCD7:ETA. In mixed culture experiments with CD7-positive and CD7-negative tumor cells, scFvCD7:sTRAIL induced very potent bystander apoptosis of CD7-negative tumor cells. In vitro treatment of blood cells freshly derived from T-acute lymphoblastic leukemia patients resulted in marked apoptosis of the malignant T cells that was strongly augmented by vincristin. In conclusion, scFvCD7:sTRAIL is a novel recombinant protein causing restricted apoptosis in human leukemic T cells with low toxicity for normal human blood and endothelial cells.


Subject(s)
Antigens, CD7/immunology , Apoptosis/drug effects , Immunotoxins/pharmacology , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Membrane Glycoproteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD7/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins , CHO Cells , Cell Line, Tumor , Cricetinae , Drug Synergism , Epitopes , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/pharmacology , Immunotoxins/genetics , Immunotoxins/immunology , Jurkat Cells/cytology , Jurkat Cells/drug effects , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/pathology , Membrane Glycoproteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/genetics , Vincristine/pharmacology
19.
Biomaterials ; 26(23): 4874-81, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15763267

ABSTRACT

The foreign body response is characterized by enhanced recruitment of inflammatory cells. As the directional movement of cells is controlled by chemokines, disruption of the chemokine network would be an attractive approach to improve biocompatibility of an implanted material. The sequestration of chemokines by cell surface-expressed glycosaminoglycans (GAGs) is vital for in vivo chemokine activity. The myxoma virus encodes a soluble protein, M-T7, that interacts with conserved GAG-binding domains of chemokines to block chemokine-mediated leukocyte recruitment. We hypothesized that M-T7 might also affect the function of other inflammation-associated proteins in addition to chemokines that bind to GAG. In our studies, we focussed on the modulation of the GAG-binding molecules macrophage chemoattractant protein-1 (MCP-1) and vascular endothelial growth factor-164 (VEGF164) in the inflammatory reaction against subcutaneously implanted degradable cross-linked dermal sheep collagen discs in AO rats. Genetic delivery of M-T7 delays the influx of macrophages into the collagen discs. In addition, angiogenesis around the implanted material was reduced. The discs revealed reduced levels of rat MCP-1 and rat VEGF164. This was not due to down regulation of transcription of the genes that encode MCP-1 and VEGF164. Our in vivo observations suggest that, in addition to chemokines such as MCP-1, M-T7 neutralizes VEGF164.


Subject(s)
Foreign-Body Reaction/immunology , Foreign-Body Reaction/prevention & control , Genetic Therapy/methods , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/prevention & control , Receptors, Interferon/immunology , Vascular Endothelial Growth Factor A/immunology , Viral Proteins/immunology , Animals , Cell Line , Foreign-Body Reaction/pathology , Immunologic Factors/genetics , Immunologic Factors/immunology , Kidney , Male , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Rabbits , Rats , Receptors, Interferon/genetics , Transfection/methods , Vascular Endothelial Growth Factor A/genetics , Viral Proteins/genetics
20.
Cytokine Growth Factor Rev ; 16(1): 91-103, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15733834

ABSTRACT

The chemokine network is an extensive system that regulates many immune functions such as leukocyte locomotion, T cell differentiation, angiogenesis and mast cell degranulation. Tight control of chemokines is vital for proper immune function. Not surprisingly, viruses have found ways to subvert or exploit the immune system in order to persist in co-existence with their hosts. Several viral immune evasion genes encode proteins that modulate the chemokine network. We attempt to identify which aspects of the chemokine control mechanisms are susceptible to modulation. Chemokine-glycosaminoglycan interaction, extracellular processing of chemokines and chemokine scavenging will be discussed in the light of poxvirus and herpesvirus immune evasion. Viral chemokine-modulatory proteins may either be targets for anti-viral therapy or lead the way to new anti-inflammatory chemokine-modulating drugs.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Chemokines/physiology , Viral Proteins/antagonists & inhibitors , Viral Proteins/therapeutic use , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , Chemokines/antagonists & inhibitors , Herpesviridae/pathogenicity , Poxviridae/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...