Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Pharmacol ; 173(1): 103-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26398585

ABSTRACT

BACKGROUND AND PURPOSE: Most of the cases of neuromyelitis optica (NMO) are characterized by the presence of an autoantibody, NMO-IgG, which recognizes the extracellular domains of the water channel, aquaporin-4. Binding of NMO-IgG to aquaporin-4 expressed in end-feet of astrocytes leads to complement-dependent disruption of astrocytes followed by demyelination. One therapeutic option for NMO is to prevent the binding of NMO-IgG to aquaporin-4, using high-avidity, non-pathogenic-chimeric, monoclonal antibodies to this water channel. We describe here the development of such antibodies. EXPERIMENTAL APPROACH: cDNAs encoding variable regions of heavy and light chains of monoclonal antibodies against the extracellular domains of human aquaporin-4 were cloned from hybridoma total RNA and fused to those encoding constant regions of human IgG1 and Igκ respectively. Then mammalian expression vectors were constructed to establish stable cell lines secreting mature chimeric antibodies. KEY RESULTS: Original monoclonal antibodies showed high avidity binding to human aquaporin-4, as determined by ELISA. Live imaging using Alexa-Fluor-555-labelled antibodies revealed that the antibody D15107 more rapidly bound to cells expressing human aquaporin-4 than others and strongly enhanced endocytosis of this water channel, while D12092 also bound rapidly to human aquaporin-4 but enhanced endocytosis to a lesser degree. Chimeric D15107 prevented complement-dependent cytotoxicity induced by NMO-IgG from patient sera in vitro. CONCLUSIONS AND IMPLICATIONS: We have established non-pathogenic, high-avidity, chimeric antibodies against the extracellular domains of human aquaporin-4, which provide a novel therapeutic option for preventing the progress and recurrence of NMO/NMO spectrum disorders.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Affinity , Aquaporin 4/chemistry , Aquaporin 4/immunology , Autoantibodies/immunology , Neuromyelitis Optica/immunology , Animals , Astrocytes/immunology , CHO Cells , Cell Line , Cricetinae , Cricetulus , Endocytosis/immunology , Humans , Immunoglobulin G/immunology , Protein Binding , Protein Structure, Tertiary
2.
Mol Ther ; 18(6): 1173-82, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20332767

ABSTRACT

Gene therapy for cystic fibrosis (CF) is making encouraging progress into clinical trials. However, further improvements in transduction efficiency are desired. To develop a novel gene transfer vector that is improved and truly effective for CF gene therapy, a simian immunodeficiency virus (SIV) was pseudotyped with envelope proteins from Sendai virus (SeV), which is known to efficiently transduce unconditioned airway epithelial cells from the apical side. This novel vector was evaluated in mice in vivo and in vitro directed toward CF gene therapy. Here, we show that (i) we can produce relevant titers of an SIV vector pseudotyped with SeV envelope proteins for in vivo use, (ii) this vector can transduce the respiratory epithelium of the murine nose in vivo at levels that may be relevant for clinical benefit in CF, (iii) this can be achieved in a single formulation, and without the need for preconditioning, (iv) expression can last for 15 months, (v) readministration is feasible, (vi) the vector can transduce human air-liquid interface (ALI) cultures, and (vii) functional CF transmembrane conductance regulator (CFTR) chloride channels can be generated in vitro. Our data suggest that this lentiviral vector may provide a step change in airway transduction efficiency relevant to a clinical programme of gene therapy for CF.


Subject(s)
Cystic Fibrosis/therapy , Genetic Therapy , Genetic Vectors , Lentivirus/genetics , Sendai virus/genetics , Viral Envelope Proteins/genetics , Animals , Cell Differentiation , Cell Line , Female , Humans , Mice , Mice, Inbred C57BL , Transduction, Genetic
3.
Mol Ther ; 16(8): 1359-65, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18523449

ABSTRACT

Platelets are receiving much attention as novel target cells to secrete a coagulation factor for hemophilia gene therapy. In order to extend the application of platelet-directed gene therapy, we examined whether ectopic expression of activated factor VII (FVIIa) in platelets would result in an efficient bypass therapy to induce sufficient thrombin generation on platelet surfaces in mice with hemophilia A. Transduction of bone marrow cells with a simian immunodeficiency virus (SIV)-based lentiviral vector harboring the platelet-specific GPIb alpha promoter resulted in efficient transgene expression in platelets. FVIIa antigen was expressed in platelets by this SIV system; FVII transgene products were found to localize in the cytoplasm and translocate toward the sub-membrane zone and cell surface after activation. Although FVII antigen levels in platelets did not reach the therapeutic levels seen with FVIIa infusion therapy, whole-blood coagulation, as assessed by thromboelastography, was significantly improved in mice with hemophilia A. Further, we observed correction of the bleeding phenotype in mice with hemophilia A after transplantation, even in the presence of FVIII-neutralizing antibodies. Our results demonstrate that FVIIa-expressing platelets can strengthen hemostatic function and may be useful in treating hemophilia and other inherited bleeding disorders. These findings are comparable to the proven therapeutic effects of FVIIa infusion.


Subject(s)
Blood Platelets/metabolism , Factor VIIa/physiology , Hemophilia A/therapy , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , Blood Platelets/cytology , Blood Platelets/ultrastructure , Cells, Cultured , Factor VIII/immunology , Factor VIIa/genetics , Factor VIIa/metabolism , Gene Expression/drug effects , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hemophilia A/genetics , Hemophilia A/pathology , Humans , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Phenotype , Simian Immunodeficiency Virus/genetics
4.
FASEB J ; 20(9): 1522-4, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16723382

ABSTRACT

Platelets release several mediators that modify vascular integrity and hemostasis. In the present study, we developed a technique for efficient transgene expression in platelets in vivo and examined whether this targeted-gene-product delivery system using a platelet release reaction could be exploited for clinical applications. Analysis of luciferase reporter gene constructs driven by platelet-specific promoters (the GPIIb, GPIbalpha, and GPVI) revealed that the GPIbalpha promoter was the most potent in the megakaryoblastic cell line UT-7/TPO and human CD34+-derived megakaryocytes. Transduction of UT-7/TPO; CD34+-derived megakaryocytes; and c-Kit+, ScaI+, and Lineage- (KSL) murine hematopoietic stem cells with a simian immunodeficiency virus (SIV)-based lentiviral vector carrying eGFP resulted in efficient, dose-dependent expression of eGFP, and the GPIbalpha promoter seemed to bestow megakaryocytic-specific expression. Transplantation of KSL cells transduced with SIV vector containing eGFP into mice showed that there was preferable expression of eGFP in platelets driven by the GPIbalpha promoter [7-11% for the cytomeglovirus (CMV) promoter, 16-27% for the GPIbalpha promoter]. Furthermore, transplantation of ex vivo-transduced KSL cells by SIV vector carrying human factorVIII (hFVIII) driven by the GPIbalpha promoter induced the production of detectable transcripts of the hFVIII gene and the hFVIII antigen in bone marrow and spleen for at least 90 days and partially corrected the hemophilia A phenotype. Platelet-targeting gene therapy using SIV vectors appears to be promising for gene therapy approaches toward not only inherited platelet diseases but also other hemorrhagic disorders such as hemophilia A.


Subject(s)
Blood Platelets/physiology , Factor VIII/genetics , Genetic Therapy/methods , Membrane Proteins/genetics , Simian Immunodeficiency Virus/genetics , Animals , Cell Differentiation , Cell Line , Fetal Blood , Genetic Vectors , Hemophilia A/genetics , Humans , Infant, Newborn , Megakaryocytes/cytology , Membrane Glycoproteins , Membrane Proteins/metabolism , Mice , Models, Biological , Platelet Glycoprotein GPIb-IX Complex , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology
5.
J Immunol ; 169(2): 778-86, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12097380

ABSTRACT

Src homology region 2 domain-containing phosphatase 1 (SHP-1) is a key mediator in lymphocyte differentiation, proliferation, and activation. We previously showed that B cell linker protein (BLNK) is a physiological substrate of SHP-1 and that B cell receptor (BCR)-induced activation of c-Jun NH(2)-terminal kinase (JNK) is significantly enhanced in cells expressing a form of SHP-1 lacking phosphatase activity (SHP-1-C/S). In this study, we confirmed that SHP-1 also exerts negative regulatory effects on JNK activation in splenic B cells. To further clarify the role of SHP-1 in B cells, we examined how dephosphorylation of BLNK by SHP-1 affects downstream signaling events. When a BLNK mutant (BLNK Delta N) lacking the NH(2)-terminal region, which contains four tyrosine residues, was introduced in SHP-1-C/S-expressing WEHI-231 cells, the enhanced JNK activation was inhibited. Among candidate proteins likely to regulate JNK activation through BLNK, Nck adaptor protein was found to associate with tyrosine-phosphorylated BLNK and this association was more pronounced in SHP-1-C/S-expressing cells. Furthermore, expression of dominant-negative forms of Nck inhibited BCR-induced JNK activation. Finally, BCR-induced apoptosis was suppressed in SHP-1-C/S-expressing cells and coexpression of Nck SH2 mutants or a dominant-negative form of SEK1 reversed this phenotype. Collectively, these results suggest that SHP-1 acts on BLNK, modulating its association with Nck, which in turn negatively regulates JNK activation but exerts a positive effect on apoptosis.


Subject(s)
Apoptosis/immunology , Carrier Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , Phosphoproteins/metabolism , Protein Tyrosine Phosphatases/physiology , Receptors, Antigen, B-Cell/physiology , Up-Regulation/immunology , src Homology Domains/immunology , Adaptor Proteins, Signal Transducing , Adjuvants, Immunologic/physiology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Carrier Proteins/physiology , Down-Regulation/immunology , Enzyme Activation/immunology , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/physiology , Oncogene Proteins/physiology , Peptide Fragments/physiology , Phosphoproteins/physiology , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Spleen/cytology , Spleen/enzymology , Spleen/immunology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...