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










Publication year range
1.
Nutr Hosp ; 32(6): 2771-6, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26667733

ABSTRACT

BACKGROUND: we successfully reconstituted the gene from group 1 allergens of dust mites, and obtained a body of shuffled genes. In order to verify the prediction on the chimeric gene, we tentatively cloned R8 into the vector that was prokaryoticly expressed, purified and assessed for its bio-activities. METHODS: the expressed product was detected by SDSPAGE and the target protein was purified. The purified protein R8 was detected by ELISA. 75 BALB/c mice were divided into 5 groups, namely PBS, rDer f1, rDer p1, R8 and asthma group. The mice were treated with dust mite allergens at 0, 7, 14 day by intraperitoneal injection and inhaled challenge as aerosol on day 21 for 7 days. Specific allergen immunotherapy was performed using rDer f1, rDer p1 and R8 allergens respectively. The level of IFN and IL- 4 in BALF was detected by ELISA. RESULTS: the chimeric gene R8 was expressed with a band of approximately Mr 35000. Compared with groups of rDer f 1 and rDer p 1 [(80.44±15.50) and (90.79±10.38) µg/ml respectively], IgE binding capacity of the protein R8 (37.03±12.46) µg/ml was statistically lower (P<0.001). The level of IFN in sera of R8 group [(343.43±38.79) pg/ml] was higher than that of the PBS and asthma groups [(393.93±50.68) and (208.44±46.11) pg/ml respectively] (P<0.01), but no statistical difference to that of the rDer f 1 and rDer p 1 groups (P>0.05). IL-4 level in R8 group was lower markedly than the others (P<0.05 or P<0.01). CONCLUSIONS: chimeric protein R8 derived from the group 1 allergens of dust mites has been expressed with low allergenicity and high immunogenicity.


Antecedentes: se reconstituyó con éxito el gen del grupo 1 alérgenos de los ácaros del polvo, y obtuvo un conjunto de genes barajadas. Con el fin de verificar la predicción en el gen quimérico, hemos clonado tentativamente R8 en el vector que se expresó prokaryoticly, purificó y se evaluó por sus actividades-bio. Métodos: el producto expresado se detectó por SDS-PAGE y la proteína diana se purificó. La proteína purificada R8 se detectó por ELISA. Setenta y cinco ratones BALB/ c se dividieron en 5 grupos, a saber: PBS, rDer f1, rDer p1, R8 y el grupo de asma. Los ratones fueron tratados con alérgenos de ácaros del polvo a los 0, 7, 14 días mediante inyección intraperitoneal y inhaladas desafío como aerosol en día 21 durante 7 días. La inmunoterapia específica para el alérgeno se realizó utilizando rDer f1, rDer p1 y alérgenos R8, respectivamente. El nivel de IFN e IL-4 en BALF se detectó por ELISA. Resultados: el gen quimérico R8 se expresó con una banda de aproximadamente Mr 35000. En comparación con los grupos de rDer f 1 y rDer p 1 [(80,44 ± 15,50) y (90,79 ± 10,38) µg/ml, respectivamente], la capacidad de unión a IgE de la proteína R8 (37,03 ± 12,46) µg/ml fue estadísticamente inferior (P.


Subject(s)
Allergens/genetics , Allergens/immunology , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/immunology , Pyroglyphidae/genetics , Pyroglyphidae/immunology , Animals , Antigens, Dermatophagoides/immunology , Asthma/immunology , Asthma/prevention & control , Immunoglobulin E/immunology , Mice , Mice, Inbred BALB C , Prokaryotic Cells/metabolism , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control
2.
J Biosci Bioeng ; 120(3): 268-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25732206

ABSTRACT

The development of a heterologous expression system is often a principal step in biochemical and biotechnological studies on cytochromes P450 (P450s). However, heterologous expression of eukaryotic membrane-bound P450s in Escherichia coli is still a trial-and-error process because theoretical and systematical experimental procedures have not yet been established. In this study, we generated a series of chimeric variants of fungal P450s by replacing their N-terminal domains with the N-terminal domains of other P450s and explored their potentials for heterologous expression in E. coli. Large-scale screening demonstrated that a number of fungal P450s could be expressed when their N-terminal amino acid sequences were replaced with the corresponding domain of CYP5144C1, even when the expression of the non-chimeric sequence was unpromising. Furthermore, a comprehensive screening resulted in the identification of 64 different types of chimeric partners whose N-terminal domains could potentially be used to increase the expression levels of various P450s. These findings will help to elaborate experimental strategies for high-level heterologous expression of a variety of eukaryotic membrane-bound P450s in E. coli.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Fungi/enzymology , Fungi/genetics , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/genetics , Cytochrome P-450 Enzyme System/chemistry , Mutant Chimeric Proteins/chemistry , Protein Structure, Tertiary/genetics
3.
Int J Cancer ; 133(10): 2483-92, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23661285

ABSTRACT

Converting T cells into tumor cell killers by grafting them with a chimeric antigen receptor (CAR) has shown promise as a cancer immunotherapeutic. However, the inability of these cells to actively migrate and extravasate into tumor parenchyma has limited their effectiveness in vivo. Here we report the construction of a CAR containing an echistatin as its targeting moiety (eCAR). As echistatin has high binding affinity to αvß3 integrin that is highly expressed on the surface of endothelial cells of tumor neovasculature, T cells engrafted with eCAR (T-eCAR) can efficiently lyse human umbilical vein endothelial cells and tumor cells that express αvß3 integrin when tested in vitro. Systemic administration of T-eCAR led to extensive bleeding in tumor tissues with no evidence of damage to blood vessels in normal tissues. Destruction of tumor blood vessels by T-eCAR significantly inhibited the growth of established bulky tumors. Moreover, when T-eCAR was codelivered with nanoparticles in a strategically designed temporal order, it dramatically increased nanoparticle deposition in tumor tissues, pointing to the possibility that it may be used together with nanocarriers to increase their capability to selectively deliver antineoplastic drugs to tumor tissues.


Subject(s)
Melanoma, Experimental/blood supply , Melanoma, Experimental/therapy , Nanoparticles/administration & dosage , T-Lymphocytes/physiology , Amino Acid Sequence , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Blood Vessels/pathology , Cell Line , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells/pathology , Humans , Integrin alphaVbeta3/metabolism , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/genetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Antigen/metabolism , T-Lymphocytes/immunology
4.
FEBS J ; 279(2): 223-33, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22074399

ABSTRACT

We previously reported the construction and activity of a humanized, bispecific diabody (hEx3) that recruited T cells towards an epidermal growth factor receptor (EGFR) positive tumor. Herein, we describe the construction of a second functional, fully humanized, anti-EGFR bispecific diabody that recruits another subset of lymphocyte effectors, the natural killer cells, to EGFR-expressing tumor cells. After we confirmed that an anti-EGFR × anti-CD16 bispecific diabody (Ex16) consisting of a previously humanized anti-EGFR variable fragment (Fv) and a mouse anti-CD16 Fv had growth inhibitory activity, we designed a humanized anti-CD16 Fv to construct the fully humanized Ex16 (hEx16). However, the humanized form had lower activity for inhibition of cancer growth. To restore its growth inhibitory activity, we introduced mutations into the Vernier zone, which is located near the complementarity-determining regions and is involved in their binding activity. We efficiently prepared 15 different hEx16 mutants by expressing each chimeric single-chain component for hEx16 separately. We then used our in vitro refolding system to select the most functional mutant, which had a growth inhibitory effect comparable with that of the commercially available chimeric anti-EGFR antibody, cetuximab. Our refolding system could aid in the efficient optimization of other proteins with heterodimeric structure.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Monoclonal, Humanized/biosynthesis , Drug Design , ErbB Receptors/antagonists & inhibitors , Immunologic Factors/metabolism , Receptors, IgG/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Coculture Techniques , ErbB Receptors/metabolism , GPI-Linked Proteins/antagonists & inhibitors , Humans , Hybridomas , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Mice , Molecular Sequence Data , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/chemistry , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Protein Refolding , Sequence Alignment
5.
J Immunol ; 186(5): 3215-25, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21289302

ABSTRACT

Antitumor alkylphospholipids initiate apoptosis in transformed HL-60 and Jurkat cells while sparing their progenitors. 1-O-Alkyl-2-carboxymethyl-sn-glycero-3-phosphocholine (Edelfosine) like other short-chained phospholipids--inflammatory platelet-activating factor (PAF) and apoptotic oxidatively truncated phospholipids--are proposed to have intracellular sites of action, yet a conduit for these choline phospholipids into mammalian cells is undefined. Edelfosine is also accumulated by Saccharomyces cerevisiae in a process requiring the membrane protein Lem3p, and the human genome contains a Lem3p homolog TMEM30a. We show that import of choline phospholipids into S. cerevisiae ΔLem3 is partially reconstituted by human TMEM30a and by Lem3p-TMEM30a chimeras, showing the proteins are orthologous. TMEM30a-GFP chimeras expressed in mammalian cells localized in plasma membranes, as well as internal organelles, and ectopic TMEM30a expression promoted uptake of exogenous choline and ethanolamine phospholipids. Short hairpin RNA knockdown of TMEM30a reduced fluorescent choline phospholipid and [(3)H]PAF import. This knockdown also reduced mitochondrial depolarization from exogenous Edelfosine or the mitotoxic oxidatively truncated phospholipid azelaoyl phosphatidylcholine, and the knockdown reduced apoptosis in response to these two phospholipids. These results show that extracellular choline phospholipids with short sn-2 residues can have intracellular roles and sites of metabolism because they are transport substrates for a TMEM30a phospholipid import system. Variation in this mechanism could limit sensitivity to short chain choline phospholipids such as Edelfosine, PAF, and proapoptotic phospholipids.


Subject(s)
Antineoplastic Agents/metabolism , Choline/metabolism , Membrane Proteins/physiology , Phospholipids/metabolism , Amino Acid Sequence , Animals , Biological Transport, Active/genetics , Biological Transport, Active/immunology , CHO Cells , Cell Line, Transformed , Cricetinae , Cricetulus , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , HL-60 Cells , Hep G2 Cells , Humans , Jurkat Cells , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , Molecular Sequence Data , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Sequence Homology, Amino Acid
6.
Brain Res ; 1351: 1-12, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20599821

ABSTRACT

Because of the heterogeneous cellular composition of the brain, and especially the forebrain, cell type-specific expression will benefit many potential applications of direct gene transfer. The two prevalent approaches for achieving cell type-specific expression are using a cell type-specific promoter or targeting gene transfer to a specific cell type. Targeted gene transfer with Herpes Simplex Virus (HSV-1) vectors modifies glycoprotein C (gC) to replace the heparin binding domain, which binds to many cell types, with a binding activity for a specific cell surface protein. We previously reported targeted gene transfer to nigrostriatal neurons using chimeric gC-glial cell line-derived neurotrophic factor or gC-brain-derived neurotrophic factor protein. Unfortunately, this approach is limited to cells that express the cognate receptor for either neurotrophic factor. Thus, a general strategy for targeting gene transfer to many different types of neurons is desirable. Antibody-mediated targeted gene transfer has been developed for targeting specific virus vectors to specific peripheral cell types; a specific vector particle protein is modified to contain the Staphylococcus A protein ZZ domain, which binds immunoglobulin (Ig) G. Here, we report antibody-mediated targeted gene transfer of HSV-1 vectors to a specific type of forebrain neuron. We constructed a chimeric gC-ZZ protein, and showed this protein is incorporated into vector particles and binds Ig G. Complexes of these vector particles and an antibody to the NMDA receptor NR1 subunit supported targeted gene transfer to NR1-containing neocortical neurons in the rat brain, with long-term (2 months) expression.


Subject(s)
Antibodies, Viral/genetics , Gene Transfer Techniques , Herpesvirus 1, Human/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Staphylococcal Protein A/genetics , Viral Envelope Proteins/genetics , Animals , Antibodies, Viral/biosynthesis , Cell Line , Gene Targeting/methods , Genetic Vectors , Helper Viruses/genetics , Helper Viruses/metabolism , Herpesvirus 1, Human/metabolism , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Male , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/genetics , Neocortex/physiology , Neurons/physiology , Rats , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/biosynthesis , Simplexvirus/genetics , Simplexvirus/metabolism , Staphylococcal Protein A/biosynthesis , Viral Envelope Proteins/biosynthesis
7.
J Immunol ; 184(8): 4284-94, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20220093

ABSTRACT

We investigated relationships among chimeric TCR (cTCR) expression density, target Ag density, and cTCR triggering to predict lysis of target cells by cTCR(+) CD8(+) T human cells as a function of Ag density. Triggering of cTCR and canonical TCR by Ag could be quantified by the same mathematical equation, but cTCR represented a special case in which serial triggering was abrogated. The magnitude of target lysis could be predicted as a function of cTCR triggering, and the predicted minimum cTCR density required for maximal target lysis by CD20-specific cTCR was experimentally tested. cTCR density below approximately 20,000 cTCR/cell impaired target lysis, but increasing cTCR expression above this density did not improve target lysis or Ag sensitivity. cTCR downmodulation to densities below this critical minimum by interaction with Ag-expressing targets limited the sequential lysis of targets in a manner that could be predicted based on the number of cTCRs remaining. In contrast, acute inhibition of lysis of primary, intended targets (e.g., leukemic B cells) due to the presence of an excess of secondary targets (e.g., normal B cells) was dependent on the Ag density of the secondary target but occurred at Ag densities insufficient to promote significant cTCR downmodulation, suggesting a role for functional exhaustion rather than insufficient cTCR density. This suggests increasing cTCR density above a critical threshold may enhance sequential lysis of intended targets in isolation, but will not overcome the functional exhaustion of cTCR(+) T cells encountered in the presence of secondary targets with high Ag density.


Subject(s)
Cytotoxicity, Immunologic/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Models, Immunological , Mutant Chimeric Proteins/antagonists & inhibitors , Mutant Chimeric Proteins/genetics , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/genetics , Antigens, CD20/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/immunology , Dose-Response Relationship, Immunologic , Humans , Jurkat Cells , Linear Models , Mutant Chimeric Proteins/biosynthesis , Predictive Value of Tests , Receptors, Antigen, T-Cell/biosynthesis , Sialic Acid Binding Ig-like Lectin 2/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
8.
Biochemistry ; 49(9): 1943-53, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20102159

ABSTRACT

The cofactor-binding domains (residues 153-295) of the alcohol dehydrogenases from the thermophile Thermoanaerobacter brockii (TbADH), the mesophilic bacterium Clostridium beijerinckii (CbADH), and the protozoan parasite Entamoeba histolytica (EhADH1) have been exchanged. Three chimeras have been constructed. In the first chimera, the cofactor-binding domain of thermophilic TbADH was replaced with the cofactor-binding domain of its mesophilic counterpart CbADH [chimera Chi21((TCT))]. This domain exchange significantly destabilized the parent thermophilic enzyme (DeltaT(1/2) = -18 degrees C). The reverse exchange in CbADH [chimera Chi22((CTC))], however, had little effect on the thermal stability of the parent mesophilic protein. Furthermore, substituting the cofactor-binding domain of TbADH with the homologous domain of EhADH1 [chimera Chi23((TET))] substantially reduced the thermal stability of the thermophilic ADH (DeltaT(1/2) = -51 degrees C) and impeded the oligomerization of the enzyme. All three chimeric proteins and one of their site-directed mutants were crystallized, and their three-dimensional (3D) structures were determined. Comparison of the 3D structures of the chimeras and the chimeric mutant with the structures of their parent ADHs showed no significant changes to their Calpha chains, suggesting that the difference in the thermal stability of the three parent ADHs and their chimeric mutants could be due to a limited number of substitutions located at strategic positions, mainly at the oligomerization interfaces. Indeed, stabilization of the chimeras was achieved, to a significant extent, either by introduction of a proline residue at a strategic position in the major horse liver ADH-type dimerization interface (DeltaT(1/2) = 35 degrees C) or by introduction of intersubunit electrostatic interactions (DeltaT(1/2) = 6 degrees C).


Subject(s)
Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Bacterial Proteins/chemistry , Clostridium beijerinckii/enzymology , Entamoeba histolytica/enzymology , Mutant Chimeric Proteins/chemistry , Protozoan Proteins/chemistry , Thermoanaerobacter/enzymology , Alcohol Oxidoreductases/biosynthesis , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Clostridium beijerinckii/genetics , Crystallography, X-Ray , Entamoeba histolytica/genetics , Enzyme Stability/genetics , Mutagenesis, Site-Directed , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/genetics , Protein Structure, Quaternary/genetics , Protein Structure, Tertiary/genetics , Protozoan Proteins/biosynthesis , Protozoan Proteins/genetics , Temperature , Thermoanaerobacter/genetics
9.
J Neurochem ; 112(1): 103-11, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19840217

ABSTRACT

Recently, we have shown that the alpha-helix present at the N-termini of alpha7 nicotinic acetylcholine receptors plays a crucial role in their biogenesis. Structural data suggest that this helix interacts with the loop linking beta-strands beta2 and beta3 (loop 3). We studied the role of this loop as well as its interaction with the helix in membrane receptor expression. Residues from Asp62 to Val75 in loop 3 were mutated. Mutations of conserved amino acids, such as Asp62, Leu65 and Trp67 abolished membrane receptor expression in Xenopus oocytes. Others mutations, at residues Asn68, Ala69, Ser70, Tyr72, Gly74, and Val 75 were less harmful although still produced significant expression decreases. Steady state levels of wild-type and mutant alpha7 receptors (L65A, W67A, and Y72A) were similar but the formation of pentameric receptors was impaired in the latter (W67A). Mutation of critical residues in subunits of heteromeric nicotinic acetylcholine receptors (alpha3beta4) also abolished their membrane expression. Complementarity between the helix and loop 3 was evidenced by studying the expression of chimeric alpha7 receptors in which these domains were substituted by homologous sequences from other subunits. We conclude that loop 3 and its docking to the alpha-helix is an important requirement for receptor assembly.


Subject(s)
Protein Subunits/biosynthesis , Protein Subunits/genetics , Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Cattle , Female , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/genetics , Protein Binding/genetics , Protein Structure, Secondary/genetics , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
10.
Retrovirology ; 6: 58, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19505341

ABSTRACT

BACKGROUND: The pig-tailed macaques are the only Old World monkeys known to be susceptible to human immunodeficiency virus type 1 (HIV-1) infection. We have previously reported that the TRIM5-Cyclophilin A (TRIMCyp) fusion in pig-tailed macaques (Macaca nemestrina) is dysfunctional in restricting HIV-1, which may explain why pig-tailed macaques are susceptible to HIV-1 infection. Similar results have also been reported by other groups. However, according to the current primate taxonomy, the previously reported M. nemestrina are further classified into three species, which all belong to the Macaca spp. This calls for the need to look into the previous studies in more details. RESULTS: The local species Northern pig-tailed macaque (M. leonina) was analyzed for the correlation of TRIM5 structure and HIV-1 infection. Eleven M. leonina animals were analyzed, and all of them were found to possess TRIM5-CypA fusion at the TRIM5 locus. The transcripts encoding the dysfunctional TRIM5-CypA should result from the G-to-T mutation in the 3'-splicing site of intron 6. Polymorphism in the putative TRIMCyp recognition domain was observed. The peripheral blood mononuclear cells (PBMCs) of M. leonina were susceptible to HIV-1 infection. Consistent with the previous results, expression of the M. leonina TRIMCyp in HeLa-T4 cells rendered the cells resistant to HIV-2ROD but not to SIVmac239 infection. CONCLUSION: The susceptibility of M. leonina to HIV-1 infection is due to the dysfunctional TRIM5-CypA fusion in the TRIM5 locus. This finding should broaden our perspective in developing better HIV/AIDS non-human primate animal models.


Subject(s)
Genetic Predisposition to Disease , HIV Infections/veterinary , HIV-1 , Macaca , Monkey Diseases/genetics , Mutant Chimeric Proteins/genetics , Animals , Base Sequence , DNA/analysis , DNA/genetics , Genotype , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HeLa Cells , Humans , Leukocytes, Mononuclear/virology , Molecular Sequence Data , Monkey Diseases/metabolism , Monkey Diseases/virology , Mutant Chimeric Proteins/biosynthesis , Phylogeny , Polymorphism, Single Nucleotide , Species Specificity
11.
J Neurosci Res ; 87(12): 2624-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19405107

ABSTRACT

Nervous system growth factor gene delivery can promote axonal growth and prevent cell death in animal models of CNS trauma and neurodegenerative diseases. The ability to regulate growth factor expression or signaling pathways downstream from growth factor receptors remains a desirable goal for in vivo gene transfer. To achieve precise pharmacological modulation of neurotrophin activity, we have generated a chimeric trkA receptor (ItrkA) by fusing the entire intracellular domain of the trkA high-affinity NGF receptor to two intracellular, modified FK506 binding domains for the synthetic small molecule dimerization ligand AP20187. Rat PC12 cells were transduced with lentiviral vectors containing ItrkA and green fluorescent protein (GFP; via an internal ribosome entry site). Treatment of ItrkA-expressing PC12 cells with AP20187 induced neurite outgrowth and differentiation in a time- and dose-dependent fashion, with a half-maximal response at a concentration of 1 nM AP20187. Seventy percent of cells responded to AP20187 by day 3. Western blots demonstrated that AP20187 treatment resulted in phosphorylation of Erk1/2 and Akt in ItrkA-transduced PC12 cells but not in nontransduced, naïve cells. Phosphorylation levels were comparable to levels obtained with 50 ng/ml nerve growth factor (NGF). In addition, ItrkA lentiviral transduction of primary E15 dorsal root ganglion neurons significantly increased neurite growth three- to fourfold in the presence of AP20187 compared with control GFP transduced and naïve neurons. These results demonstrate that small ligand-induced dimerization of the intracellular domain of trkA can efficiently simulate the biological activity of NGF and provide a means to regulate intracellular neurotrophin receptor signaling.


Subject(s)
Nerve Growth Factor/metabolism , Nervous System/embryology , Neurites/metabolism , Neurogenesis/physiology , Receptor, trkA/metabolism , Signal Transduction/physiology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Genetic Vectors/genetics , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mutant Chimeric Proteins/biosynthesis , Nerve Growth Factor/agonists , Nervous System/cytology , Nervous System/growth & development , Neurites/drug effects , Neurogenesis/drug effects , PC12 Cells , Phosphorylation/drug effects , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, trkA/chemistry , Receptor, trkA/genetics , Recombinant Fusion Proteins/biosynthesis , Signal Transduction/drug effects , Tacrolimus/analogs & derivatives , Tacrolimus/chemistry , Tacrolimus/pharmacology , Transduction, Genetic/methods
12.
BMC Mol Biol ; 9: 81, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18817541

ABSTRACT

BACKGROUND: Regulation of the expression of particular genes can rely on mechanisms that are different from classical transcriptional and translational control. The LY6G5B and LY6G6D genes encode LY-6 domain proteins, whose expression seems to be regulated in an original fashion, consisting of an intron retention event which generates, through an early premature stop codon, a non-coding transcript, preventing expression in most cell lines and tissues. RESULTS: The MHC LY-6 non-coding transcripts have shown to be stable and very abundant in the cell, and not subject to Nonsense Mediated Decay (NMD). This retention event appears not to be solely dependent on intron features, because in the case of LY6G5B, when the intron is inserted in the artificial context of a luciferase expression plasmid, it is fully spliced but strongly stabilises the resulting luciferase transcript. In addition, by quantitative PCR we found that the retained and spliced forms are differentially expressed in tissues indicating an active regulation of the non-coding transcript. EST database analysis revealed that these genes have an alternative expression pathway with the formation of Transcription Induced Chimeras (TIC). This data was confirmed by RT-PCR, revealing the presence of different transcripts that would encode the chimeric proteins CSNKbeta-LY6G5B and G6F-LY6G6D, in which the LY-6 domain would join to a kinase domain and an Ig-like domain, respectively. CONCLUSION: In conclusion, the LY6G5B and LY6G6D intron-retained transcripts are not subjected to NMD and are more abundant than the properly spliced forms. In addition, these genes form chimeric transcripts with their neighbouring same orientation 5' genes. Of interest is the fact that the 5' genes (CSNKbeta or G6F) undergo differential splicing only in the context of the chimera (CSNKbeta-LY6G5B or G6F-LY6G6C) and not on their own.


Subject(s)
Antigens, Ly/genetics , Introns , Mutant Chimeric Proteins/genetics , Alternative Splicing , Antigens, Ly/biosynthesis , Cell Line , Codon, Nonsense , Gene Expression Regulation , Humans , Mutant Chimeric Proteins/biosynthesis , Organ Specificity , RNA Stability
SELECTION OF CITATIONS
SEARCH DETAIL