Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 15(12): 5665-5677, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30375878

ABSTRACT

Protein replacement therapy (PRT) has been applied to treat severe monogenetic/metabolic disorders characterized by a protein deficiency. In disorders where an intracellular protein is missing, PRT is not easily feasible due to the inability of proteins to cross the cell membrane. Instead, gene therapy has been applied, although still with limited success. ß-Thalassemias are severe congenital hemoglobinopathies, characterized by deficiency or reduced production of the adult ß-globin chain. The resulting imbalance of α-/ß-globin chains of adult hemoglobin (α2ß2) leads to precipitation of unpaired α-globin chains and, eventually, to defective erythropoiesis. Since protein transduction domain (PTD) technology has emerged as a promising therapeutic approach, we produced a human recombinant ß-globin chain in fusion with the TAT peptide and successfully transduced it into human proerythroid K-562 cells, deficient in mature ß-globin chain. Notably, the produced human recombinant ß-globin chain without the TAT peptide, used as internal negative control, failed to be transduced into K-562 cells under similar conditions. In silico studies complemented by SDS-PAGE, Western blotting, co-immunoprecipitation and LC-MS/MS analysis indicated that the transduced recombinant fusion TAT-ß-globin protein interacts with the endogenous native α-like globins to form hemoglobin α2ß2-like tetramers to a limited extent. Our findings provide evidence that recombinant TAT-ß-globin is transmissible into proerythroid K-562 cells and can be potentially considered as an alternative protein therapeutic approach for ß-thalassemias.


Subject(s)
Recombinant Fusion Proteins/therapeutic use , beta-Globins/therapeutic use , beta-Thalassemia/therapy , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Biological Therapy/methods , Cell Line , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Transduction, Genetic/methods , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/isolation & purification , beta-Thalassemia/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/isolation & purification
2.
Ann Clin Lab Sci ; 48(4): 460-468, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30143487

ABSTRACT

OBJECTIVE: Excitotoxic neuronal death induced by epilepsy is associated with α-amino-3-hydroxyl-5-methylisoxazole-4-propionate acid (AMPA) receptors. The GluR2 subunit of AMPA receptors (AMPARs) may bind with glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The GluR2/GAPDH complex co-internalizes upon stimulation of AMPARs, which might be involved in the development of epilepsy. In this research, we hypothesized that disruption of the GluR2/GAPDH interaction with an interfering peptide would protect against neuronal damage in vivo. METHODS: Rat models of epilepsy were induced by pilocarpine hydrochloride. TAT-GluR2NT1-3-2 peptide was synthesized to block interaction between GluR2 and GAPDH. Fluoro-Jade B and TUNEL staining were used to detect degeneration and apoptosis of neurons after interference by the peptide. Co-immunoprecipitation assay and western-blot was performed to confirm that the peptide disturbed interactions between GluR2 and GAPDH. RESULTS: The time of epileptic seizure was found to be delayed after peptide interference. It was concluded that administration of an interfering peptide is able to significantly reduce degeneration and apoptosis of neurons. The GluR2/GAPDH interaction and GAPDH nuclear expression were upregulated in the hippocampus of rats subjected to pilocarpine-induced seizures. CONCLUSION: Disruption of the GluR2/GAPDH interaction by administration of an interfering peptide protects against seizure-induced neuronal damage that is dose dependent. Thus, the GluR2/GAPDH interaction may be a novel therapeutic target for development of treatment for epilepsy.


Subject(s)
Epilepsy/drug therapy , Epilepsy/pathology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Peptides/therapeutic use , Receptors, AMPA/metabolism , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Amino Acid Sequence , Animals , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Epilepsy/chemically induced , Hippocampus/drug effects , Hippocampus/pathology , Male , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Pilocarpine , Protein Transport/drug effects , Rats, Wistar , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/pharmacology
3.
Chem Biol Drug Des ; 92(2): 1567-1575, 2018 08.
Article in English | MEDLINE | ID: mdl-29722179

ABSTRACT

To enhance the anticancer activity, tumor penetration ability of the hybrid anticancer peptide, in this study, a TAT (RKKRRQRRR) peptide modified kla peptide (KLAKLAKKLAKLAK, with all D-amino acids), named kla-TAT, was co-administrated with the homing/penetrating peptide iRGD which could enhance the permeability of chemical drug in solid tumor and tumor vessel by co-administration. In this study, the nonsmall cell lung cancer A549 cell line with the iRGD targeting receptor neuropilin-1 high expression was selected to establish the 2D monolayer cell, 3D multiple cell spheroids, and xenograft mice model. The co-administration of iRGD strengthened the permeability of kla-TAT peptide against A549 2D and 3D sphere model with the penetration improvement property of iRGD; more importantly, co-administration with iRGD dramatically enhanced the accumulation of kla-TAT peptide in tumor tissue on the xenograft mice model with the homing property of iRGD. The co-administration of iRGD strategy confers targeting ability to the hybrid peptide kla-TAT. We believe the chemical conjugation plus co-administration approach may provide a promising way for cancer treatment in clinical practices.


Subject(s)
Oligopeptides/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry , A549 Cells , Amino Acid Sequence , Animals , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/metabolism , Oligopeptides/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Peptides/pharmacology , Peptides/therapeutic use , Permeability/drug effects , Transplantation, Heterologous , tat Gene Products, Human Immunodeficiency Virus/pharmacology , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
4.
Nanomedicine (Lond) ; 11(14): 1769-85, 2016 07.
Article in English | MEDLINE | ID: mdl-27388639

ABSTRACT

AIM: In the present study, we examine the effects of internalized peptide-conjugated iron oxide nanoparticles and their ability to locally convert alternating magnetic field (AMF) energy into other forms of energy (e.g., heat and rotational work). MATERIALS & METHODS: Dextran-coated iron oxide nanoparticles were functionalized with a cell penetrating peptide and after internalization by A549 and H358 cells were activated by an AMF. RESULTS: TAT-functionalized nanoparticles and AMF exposure increased reactive oxygen species generation compared with the nanoparticle system alone. The TAT-functionalized nanoparticles induced lysosomal membrane permeability and mitochondrial membrane depolarization, but these effects were not further enhanced by AMF treatment. Although not statistically significant, there are trends suggesting an increase in apoptosis via the Caspase 3/7 pathways when cells are exposed to TAT-functionalized nanoparticles combined with AMF. CONCLUSION: Our results indicate that internalized TAT-functionalized iron oxide nanoparticles activated by an AMF elicit cellular responses without a measurable temperature rise.


Subject(s)
Lung Neoplasms/therapy , Magnetite Nanoparticles/therapeutic use , Peptide Fragments/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lysosomes/metabolism , Lysosomes/pathology , Magnetic Fields , Magnetics , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/ultrastructure , Peptide Fragments/chemistry , Reactive Oxygen Species/metabolism , tat Gene Products, Human Immunodeficiency Virus/chemistry
5.
Int J Mol Med ; 38(1): 217-24, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27222268

ABSTRACT

Antioxidant 1 (ATOX1) functions as an antioxidant against hydrogen peroxide and superoxide, and therefore may play a significant role in many human diseases, including diabetes mellitus (DM). In the present study, we examined the protective effects of Tat-ATOX1 protein on streptozotocin (STZ)-exposed pancreatic insulinoma cells (RINm5F) and in a mouse model of STZ-induced diabetes using western blot analysis, immunofluorescence staining and MTT assay, as well as histological and biochemical analysis. Purified Tat-ATOX1 protein was efficiently transduced into RINm5F cells in a dose- and time-dependent manner. Additionally, Tat-ATOX1 protein markedly inhibited reactive oxygen species (ROS) production, DNA damage and the activation of Akt and mitogen activated protein kinases (MAPKs) in STZ-exposed RINm5F cells. In addition, Tat-ATOX1 protein transduced into mice pancreatic tissues and significantly decreased blood glucose and hemoglobin A1c (HbA1c) levels as well as the body weight changes in a model of STZ-induced diabetes. These results indicate that transduced Tat-ATOX1 protein protects pancreatic ß-cells by inhibiting STZ-induced cellular toxicity in vitro and in vivo. Based on these findings, we suggest that Tat-ATOX1 protein has potential applications as a therapeutic agent for oxidative stress-induced diseases including DM.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Metallochaperones/therapeutic use , Pancreas/pathology , Recombinant Fusion Proteins/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Animals , Cell Death/drug effects , Cell Line, Tumor , Copper Transport Proteins , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Humans , Male , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/metabolism , Molecular Chaperones , Proto-Oncogene Proteins c-akt/metabolism , Rats , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/pharmacology
6.
Hum Gene Ther ; 26(10): 680-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26077977

ABSTRACT

Today, lentiviral vectors are favorable vectors for RNA interference delivery in anti-HIV therapeutic approaches. Nevertheless, problems such as the specific recognition of target cells and uncontrolled expression of the transgene can restrict their use in vivo. Herein we present a new HIV-inducible promoter to express anti-HIV short hairpin RNA (shRNA) by RNA Pol II in mammalian cells. We likewise showed a novel third-generation lentiviral vector system with more safety and a specific tropism to the target cells. The new promoter, CkRhsp, was constructed from the chicken ß-actin core promoter with the R region of HIV-1 long terminal repeat fused upstream of minimal hsp70 promoter. This system was induced by HIV-1 Tat, and activates transcription of two shRNAs against two conserved regions of HIV-1 transcripts produced in two steps of the virus life cycle. We also mimicked HIV-1 cell tropism by using the HIV-1 envelope in structure of third-generation lentiviral vector. The new fusion promoter efficiently expressed shRNA in a Tat-inducible manner. HIV-1 replication was inhibited in transient transfection and stable transduction assays. The new viral vector infected only CD4+cells. CkRhsp promoter may be safer than other inducible promoters for shRNA-mediated gene therapies against HIV. The use of the wild envelope in the vector packaging system may provide the specific targeting T lymphocytes and hematopoietic stem cells for anti-HIV-1 therapeutic approaches in vivo.


Subject(s)
HIV Infections/genetics , HIV-1/genetics , RNA Polymerase II/genetics , Virus Replication/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Genetic Vectors , HIV Infections/therapy , HIV Infections/virology , HIV-1/pathogenicity , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/therapeutic use , Humans , Lentivirus , RNA Polymerase II/therapeutic use , RNA, Small Interfering , Transduction, Genetic , Viral Tropism/genetics , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
7.
Expert Opin Biol Ther ; 15 Suppl 1: S13-29, 2015.
Article in English | MEDLINE | ID: mdl-26096836

ABSTRACT

INTRODUCTION: Classical approaches aimed at targeting the HIV-1 envelope as well as other structural viral proteins have largely failed. The HIV-1 transactivator of transcription (Tat) is a key HIV virulence factor, which plays pivotal roles in virus gene expression, replication, transmission and disease progression. Notably, anti-Tat Abs are uncommon in natural infection and, when present, correlate with the asymptomatic state and lead to lower or no disease progression. Hence, targeting Tat represents a pathogenesis-driven intervention. AREAS COVERED: Here, we review the rationale and the translational development of a therapeutic vaccine targeting the Tat protein. Preclinical and Phase I studies, Phase II trials with Tat in anti-Tat Ab-negative, virologically suppressed highly active antiretroviral therapy-treated subjects in Italy and South Africa were conducted. The results indicate that Tat-induced immune responses are necessary to restore immune homeostasis, to block the replenishment and to reduce the size of the viral reservoir. Additionally, they may help in establishing key parameters for highly active antiretroviral therapy intensification and a functional cure. EXPERT OPINION: We propose the therapeutic setting as the most feasible to speed up the testing and comparison of preventative vaccine candidates, as the distinction lies in the use of the vaccine in uninfected versus infected subjects and not in the vaccine formulation.


Subject(s)
AIDS Vaccines/therapeutic use , HIV Infections/prevention & control , HIV-1 , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , AIDS Vaccines/immunology , Animals , Antiretroviral Therapy, Highly Active/methods , Clinical Trials as Topic/methods , Disease Progression , HIV Infections/immunology , HIV-1/immunology , Humans , tat Gene Products, Human Immunodeficiency Virus/immunology
8.
Mol Immunol ; 63(2): 355-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25239864

ABSTRACT

Reactive oxygen species (ROS) accumulation induces oxidative stress and cell damage, which then activates several signaling pathways and triggers inflammatory response. Biliverdin is a natural product of heme metabolism which is converted to bilirubin by the enzyme biliverdin reductase A (BLVRA) which also plays a role in antioxidant activity via the ROS scavenging activity of bilirubin. In this study, we examined the anti-inflammatory and anti-apoptotic effects of Tat-BLVRA protein on lipopolysaccharide (LPS)-induced inflammation in Raw 264.7 macrophage cells. Transduction of Tat-BLVRA protein into Raw 264.7 cells and mice ear tissue was tested by Western blot analysis and immunohistochemical analysis. Tat-BLVRA protein was effective in inhibiting mitogen activated protein kinases (MAPKs), Akt and NF-κB activation, intracellular ROS production and DNA fragmentation. Also, Tat-BLVRA protein significantly inhibited the expression of cytokines, COX-2, and iNOS. In a 12-O-tetradecanoylphobol 13-acetate (TPA)-induced mouse model, mice ears treated with Tat-BLVRA protein showed decreased ear thickness and weight, as well as inhibited MAPKs activation and cytokine expression. Thus we suggested that Tat-BLVRA protein may provide an effective therapeutic agent for inflammatory skin diseases.


Subject(s)
Edema/therapy , Inflammation/pathology , Macrophages/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Animals , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Edema/pathology , Humans , Inflammation/enzymology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Male , Mice, Inbred ICR , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/isolation & purification , Signal Transduction , Tetradecanoylphorbol Acetate , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/metabolism
9.
CNS Neurol Disord Drug Targets ; 13(8): 1465-74, 2014.
Article in English | MEDLINE | ID: mdl-25106625

ABSTRACT

AIMS: To compare the neuroprotection of erythropoietin (EPO) and EPO fusion protein containing transduction domain derived from HIV TAT (EPO-TAT) against ischemic brain injury, inclusive of the side effect, and explore the mechanism underlying the role of EPO-TAT in a transient focal cerebral ischemia model in rats. METHODS: Transient focal ischemia was induced by middle cerebral artery occlusion (MCAO) in rats. Rats were treated, respectively, with following regimens: saline, 1000 U/kg EPO, 5000 U/kg EPO, 1000 U/kg EPO-TAT, 1000 U/kg EPOTAT+5 µl of 10 mM LY294002 (or/plus 5 µl of 5 mM PD98059). Neurological deficit scores, infarct volume, and hematologic side effect were assessed at 72 hours after MCAO. Apoptotic cells were determined with TUNEL staining. The expression and localization of phosphorylated AKT (pAKT) and phosphorylated ERK (pERK) were detected with Western blot, immunohistochemistry, and immunofluorescence, respectively. RESULTS: 1000 U/kg EPO-TAT exhibited a comparable neuroprotection to 5000 U/kg EPO, as evidenced by a comparable attenuation in neurological deficit, infarct volume, and number of apoptotic cells in the rat ischemic cortex after MCAO. The pAKT and pERK levels were significantly elevated solely in neurons of rodents receiving EPO or EPO-TAT treatments, suggesting the concurrent activation of these two pathways. Specific inhibition of either AKT or ERK pathway partially abolished EPO-TAT protection, but exhibited no influence on the activation status of its counterpart, suggesting no cross-modulation between these two protective pathways. CONCLUSION: Our study indicates that EPO-TAT at 1000 U/kg displays neuroprotection with no detectable side effects. The mechanism for neuroprotection may be attributable to the simultaneous activation of the AKT and ERK pathways, which preserve neuronal cell viability and attenuate behavioral deficits.


Subject(s)
Erythropoietin/therapeutic use , Infarction, Middle Cerebral Artery/complications , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/therapeutic use , Animals , Cell Death/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , In Situ Nick-End Labeling , Male , Neurologic Examination , Oncogene Protein v-akt/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/therapeutic use , Statistics, Nonparametric , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
11.
CNS Neurosci Ther ; 19(5): 319-28, 2013 May.
Article in English | MEDLINE | ID: mdl-23490284

ABSTRACT

AIMS: In acute stroke, neurological damage is due to oxidative stress and neuronal apoptotic death. This study investigated whether Nogo-A 290-562 residues region (M9), fused to the transduction domain of the HIV trans-activator (TAT) protein, is neuroprotective against cerebral ischemia and the mechanisms. METHODS: Transient focal cerebral ischemia was induced by middle cerebral artery occlusion in male C57BL/6J mice. TAT-M9, its mutation or vehicle was applied via intraperitoneal injection at the onset of reperfusion. The neurobehavioral scores, infarction volumes, neuronal apoptosis, and the ratio of Bax/Bcl-2 were evaluated. Malondialdehyde (MDA), reactive oxygen species (ROS) levels, and NADPH oxidase activation were measured in the presence or absence of the NADPH oxidase inhibitor apocynin or activator tetrabromocinnamic acid (TBCA). RESULTS: Immunofluorescence results confirmed that TAT-M9 was transduced into brain parenchyma, and it significantly improved neurological behavior, reduced infarct volumes, protected neuronal cells from apoptosis, inhibited activation of NADPH oxidase, and decreased MDA and ROS contents. Furthermore, apocynin imitated the beneficial effects of TAT-M9, while TBCA abolished them. CONCLUSIONS: Our results demonstrate that TAT-M9 administration attenuates cerebral ischemia by inhibiting NADPH oxidase-mediated oxidative damage and neuronal apoptosis in mice. TAT-M9 may be a potential treatment for cerebrovascular disease.


Subject(s)
Brain Ischemia/drug therapy , Myelin Proteins/therapeutic use , NADPH Oxidases/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Superoxides/metabolism , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Animals , Apoptosis/drug effects , Brain Ischemia/metabolism , Brain Ischemia/pathology , Mice , Mice, Inbred C57BL , NADPH Oxidases/physiology , Nogo Proteins
12.
J Neurotrauma ; 28(7): 1219-28, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21510821

ABSTRACT

Astrocyte activation contributes to the brain's response to disease and injury. Activated astrocytes generate harmful radicals that exacerbate brain damage including nitric oxide, peroxides and superoxides. Furthermore, reactive astrocytes hinder regeneration of damaged neural circuits by secreting neuro-developmental inhibitors and glycosaminoglycans (GAGs), which physically block growth cone extension. Therefore, targeted therapeutic strategies to limit astrocyte activation may enhance recovery from many neurodegenerative states. Previously, we demonstrated that the HIV-1 TAT cell-penetrating peptide, a short non-toxic peptide from the full-length TAT protein, delivered a protein cargo to astrocytes in a process dependent on cell-surface GAG. Since activated astrocytes produce GAG, in this study we tested whether TAT could transduce activated astrocytes, deliver a biologically active cargo, and produce a physiological effect. Astrocyte activation was induced by IL-1ß, lipopolysaccharide (LPS), or mechanical stretch injury, and quantified by increased GAG and nitrite content. TAT-mediated delivery of a mock therapeutic protein, GFP, increased significantly after activation. Nitrite production, GAG expression, and GFP-TAT transduction were significantly attenuated by inhibitors of JNK, p38, or ERK. TAT fused to a peptide JNK inhibitor delivered the peptide inhibitor to activated astrocytes and significantly reduced activation. Our study is the first to report significant and direct modulation of astrocyte activation with a peptide JNK inhibitor. Our promising in vitro results warrant in vivo follow-up, as TAT-mediated protein delivery may have broad therapeutic potential for preventing astrocyte activation with the possibility of limiting off-target, negative side effects.


Subject(s)
Astrocytes/enzymology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Peptides/physiology , Protein Kinase Inhibitors/pharmacology , tat Gene Products, Human Immunodeficiency Virus/physiology , Animals , Astrocytes/drug effects , Astrocytes/physiology , Biological Transport, Active/physiology , Drug Delivery Systems/methods , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gliosis/drug therapy , Gliosis/enzymology , Gliosis/etiology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Peptides/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , Transduction, Genetic , Treatment Outcome , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
13.
Neurobiol Dis ; 40(1): 265-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20554038

ABSTRACT

Neural precursor cells (NPC) are an interesting tool in experimental stroke research, but their therapeutic potential is limited due to poor long-term survival. We therefore in vitro transduced subventricular zone-(SVZ)-derived NPC with the anti-apoptotic fusion protein TAT-Bcl-x(L) and analyzed NPC survival, differentiation, and post-stroke functional deficits after experimental ischemia in mice. Survival of TAT-Bcl-x(L)-transduced NPC, which were injected at day 7 post-stroke into the ischemic striatum, was significantly increased at 4 weeks after stroke. Increased survival of NPC was associated with reduced infarct injury and decreased post-stroke functional deficits. Animals grafted with TAT-Bcl-x(L)-transduced NPC showed an increased number of immature cells expressing the neuronal marker doublecortin. Since mature neuronal differentiation of NPC was not observed, reduced post-stroke injury cannot be attributed to enhanced neuronal regeneration, but rather to indirect by-stander effects of grafted NPC. In line with this, NPC-mediated neuroprotection of cortical neurons in vitro was associated with increased secretion of growth factors. Thus, in vitro transduction of cultivated NPC with TAT-Bcl-x(L) results in enhanced resistance of transplanted NPC followed by long-term neuroprotection and ameliorated functional deficits after transient focal cerebral ischemia in mice.


Subject(s)
Brain Ischemia/genetics , Brain Ischemia/therapy , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Stroke/therapy , Transduction, Genetic/methods , bcl-X Protein/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/surgery , Cell Survival/genetics , Cytoprotection/genetics , Disease Models, Animal , Gene Transfer Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Recovery of Function/genetics , Stroke/genetics , Stroke/pathology , Time , bcl-X Protein/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
14.
Cancer Sci ; 101(1): 87-93, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19804421

ABSTRACT

Interaction of the Eph family of receptor protein tyrosine kinases and their ligands, ephrin family members, induces bidirectional signaling through cell-cell contacts. High expression of B-type ephrin is associated with high invasion potential of tumors, and we previously observed that signaling through the C-terminus of ephrin-B1 mediates the migration and invasion of cells, and is involved in the promotion of carcinomatous peritonitis in vivo. Here we show that the intracellular introduction of a synthetic peptide derived from ephrin-B1 C-terminus blocks ephrin-B1 mediated signaling in scirrhous gastric cancer cells. Treatment of cancer cells with a fusion peptide consisting of HIV-TAT and amino acids 331-346 of ephrin-B1 (PTD-EFNB1-C) suppressed the activation of RhoA, mediated by the association of ephrin-B1 with an adaptor protein Dishevelled, and also inhibited extracellular secretion of metalloproteinase. Moreover, injection of PTD-EFNB1-C peptide into the peritoneal cavity of nude mice suppressed carcinomatous peritonitis of intraperitoneally transplanted scirrhous gastric cancer cells. These results indicate the possible application of ephrin-B1 C-terminal peptide to develop novel protein therapy for scirrhous gastric carcinoma, especially in the stage of tumor progression, including peritoneal dissemination.


Subject(s)
Apoptosis Regulatory Proteins/therapeutic use , Ephrin-B1/antagonists & inhibitors , Peptide Fragments/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Stomach Neoplasms/drug therapy , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Amino Acid Sequence , Animals , Cell Line, Tumor , Humans , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peritoneal Neoplasms/prevention & control , Stomach Neoplasms/pathology , Survivin , rhoA GTP-Binding Protein/antagonists & inhibitors
15.
Lab Invest ; 90(2): 156-67, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20010851

ABSTRACT

Increasing evidence suggests that apoptosis may be the mechanism underlying cell death in selective loss of nigral dopaminergic neurons in Parkinson's disease (PD). Previous studies strongly suggested that c-Jun N-terminal kinase (JNK) signaling pathway has a critical role in the animal model with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD. In this study, we report the inhibitory effect of a peptide designated as Tat-JBD on JNKs activation. The sequence of Tat is corresponding to the cell-membrane transduction domain of human immunodeficiency virus-type 1 (HIV-1) and the sequence of an 11-amino acid peptide is corresponding to the residues of JNK-binding domain (JBD) on JNK-interacting protein-1 (JIP-1). Tat-JBD is confirmed to perturb the assembly of JIP-1-JNKs complex, inhibit the activation of JNKs induced by MPTP and consequently diminish the phosphorylation of c-Jun. It also inhibits the phosphorylation of Bcl-2 and the releasing of Bax from Bcl-2/Bax dimmers, sequentially attenuates the translocation of Bax to mitochondria, the release of cytochrome c, the activation of caspase3 and the hydrolyzation of poly-ADP-ribose-polymerase. The death of dopaminergic neurons and the loss of dopaminergic axon in the striatum were significantly suppressed by infusion of the peptide Tat-JBD in MPTP-treated mice. Our findings imply that Tat-JBD offers neuroprotection against MPTP injury via inhibiting the JNK-signaling pathway, and may provide a promising therapeutic approach for PD.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/drug effects , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Peptides/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Apoptosis/drug effects , Disease Models, Animal , In Situ Nick-End Labeling , Male , Mice , Nerve Degeneration/prevention & control , Phosphorylation/drug effects , Protein Kinase Inhibitors , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
16.
Appl Radiat Isot ; 67(7-8 Suppl): S88-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19446462

ABSTRACT

We aimed at securing sufficient concentrations of (10)B in boron neutron capture therapy (BNCT) by developing a new drug delivery system. We have designed and developed a novel lipid analog and succeeded in using it to develop the new boron component liposome. It consisted of three different kinds of amino acid derivatives and two fatty acids, and could react directly with the peptide synthesized first on resin by Fmoc solid-phase synthesis. In this study, lipid analog conjugated with HIV-TAT peptide (domain of human immunodeficiency virus TAT protein) and boronophenylalanine (BPA) was synthesized and successfully incorporated into liposomes.


Subject(s)
Boron Compounds/chemical synthesis , Boron Neutron Capture Therapy/methods , Liposomes/chemistry , Phenylalanine/analogs & derivatives , Radiation-Sensitizing Agents/chemical synthesis , tat Gene Products, Human Immunodeficiency Virus/chemical synthesis , Boron Compounds/chemistry , Boron Compounds/therapeutic use , Drug Delivery Systems , Humans , Isotopes/chemistry , Isotopes/therapeutic use , Lipopeptides/chemical synthesis , Lipopeptides/chemistry , Lipopeptides/therapeutic use , Membrane Potentials , Neoplasms/radiotherapy , Particle Size , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Phenylalanine/therapeutic use , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
17.
J Neurochem ; 109(6): 1680-90, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19457136

ABSTRACT

Ciliary neurotrophic factor (CNTF) is a multifunctional cytokine that can regulate the survival and differentiation of many types of developing and adult neurons. CNTF prevents the degeneration of motor neurons after axotomy and in mouse mutant progressive motor neuronopathy, which has encouraged trials of CNTF for human motor neuron disease. Given systemically, however, CNTF causes severe side effects, including cachexia and a marked immune response, which has limited its clinical application. The present work describes a novel approach for administering recombinant human CNTF (rhCNTF) while conserving neurotrophic activity and avoiding deleterious side effects. rhCNTF was fused to a protein transduction domain derived from the human immunodeficiency virus-1 TAT (transactivator) protein. The resulting fusion protein (TAT-CNTF) crosses the plasma membrane within minutes and displays a nuclear localization. TAT-CNTF was equipotent to rhCNTF in supporting the survival of cultured chicken embryo dorsal root ganglion neurons. Local or subcutaneous administration of TAT-CNTF, like rhCNTF rescued motor neurons from death in neonatal rats subjected to sciatic nerve transection. In contrast to subcutaneous rhCNTF, which caused a 20-30% decrease in body weight in neonatal rats between postnatal days 2 and 7 together with a considerable fat mobilization in brown adipose tissue, TAT-CNTF lacked such side effects. Together, these results indicate that rhCNTF fused with the protein transduction domain/TAT retains neurotrophic activity in the absence of CNTFs cytokine-like side effects and may be a promising candidate for the treatment of motor neuron and other neurodegenerative diseases.


Subject(s)
Ciliary Neurotrophic Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Sciatic Neuropathy/drug therapy , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Animals, Newborn , Axotomy/methods , Body Weight/drug effects , Cell Count/methods , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor/metabolism , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Humans , Motor Neurons/drug effects , Motor Neurons/physiology , Rats , Rats, Wistar , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , STAT3 Transcription Factor/metabolism , Sciatic Neuropathy/etiology , Sciatic Neuropathy/physiopathology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Transduction, Genetic/methods , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
18.
Med Sci (Paris) ; 24(10): 821-7, 2008 Oct.
Article in French | MEDLINE | ID: mdl-18950577

ABSTRACT

Discovered 10 years ago, survivin has a dual role in the smooth progress of mitosis and in apoptosis resistance. Survivin plays an important physiological role in development, but is absent in differentiated adult tissues. In contrast, aberrant survivin expression is found in most human cancers because of the activation of various signalling pathways. A complex survivin network appears to intersect multiple pathways in cell biology, related to several molecular partners and fine subcellular localizations. Based on its pro-oncogenic properties, basic and translational studies have shown a growing interest in survivin that has led to consider survivin as a prognostic marker and a promising target for anti-tumoral therapies.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Microtubule-Associated Proteins/physiology , Neoplasm Proteins/physiology , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Apoptosis Regulatory Proteins/therapeutic use , Biomarkers, Tumor , Cancer Vaccines/therapeutic use , Cell Cycle/physiology , Clinical Trials, Phase I as Topic , Drug Delivery Systems , Drug Screening Assays, Antitumor , Embryonic Development/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Inhibitor of Apoptosis Proteins , Models, Biological , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Recombinant Fusion Proteins/therapeutic use , Subcellular Fractions/metabolism , Survivin , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
19.
Cancer Gene Ther ; 15(5): 303-14, 2008 May.
Article in English | MEDLINE | ID: mdl-18309353

ABSTRACT

Cancer suicide gene therapy based on herpes simplex virus type I thymidine kinase (HSV-TK) and ganciclovir (GCV) suffers from the lack of efficacy in clinical use, which is mostly due to low gene-transfer efficiency and absence of bystander effect in tumors. We have previously demonstrated the enhancement of GCV cytotoxicity by fusing the HSV-TK with the cell penetrating peptide from HIV-1 transactivator protein transduction domain (TAT PTD). Despite the earlier promising results, we found that the triple fusion protein HIV-1 transactivator protein transduction domain-thymidine kinase suicide gene-green fluorescent protein marker gene (TAT-TK-GFP) increased GCV cytotoxicity only in 3/12 of different human tumor cell lines. Extended GCV exposure enhanced the cytotoxic effect of HSV-TK/GCV gene therapy, but the difference between TK-GFP and TAT-TK-GFP was not statistically significant. The modest improvement on cell killing mediated by TAT PTD in Chinese hamster ovary cells appeared to be associated with cell-surface heparan sulfate proteoglycan (HSPG) composition. However, TAT-mediated increased cell death did not correlate with the density of cell-surface HSPG expression in different tumor cell lines. In conclusion, although some degree of enhancement by TAT was shown in certain tumor cells in vitro, it is unlikely that TAT peptide linked to a suicide protein could be a useful booster of in vivo gene therapy trials.


Subject(s)
Ganciclovir/therapeutic use , Genetic Therapy/methods , Herpesvirus 1, Human/genetics , Neoplasms/genetics , Neoplasms/therapy , Peptide Fragments/therapeutic use , Thymidine Kinase/genetics , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Animals , Antiviral Agents/therapeutic use , CHO Cells , Cell Death/drug effects , Cricetinae , Cricetulus , Humans , Thymidine Kinase/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/chemistry
20.
Cancer Sci ; 99(5): 1034-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18294279

ABSTRACT

Although dendritic cells (DC) have been well demonstrated as a strong cellular adjuvant for a tumor vaccine, there are several limitations for clinical application. A protein-based vaccine using a potent adjuvant is an appealing approach for tumor antigen-specific immunotherapy because of their simplicity, safety, efficacy and capacity for repeated administration. CpG-oligodeoxynucleotides (ODN) have been used as adjuvants to stimulate innate and adaptive immune responses for cancer treatment. The authors evaluated the adjuvant effects of CpG-ODN in a vaccine incorporating recombinant fusion protein of the HIV TAT PTD domain and carcinoembryonic antigen (TAT-CEA). Mice vaccinated with TAT-CEA and CpG-ODN (TAT-CEA + CpG) showed enhanced CEA-specific immunity, including cytotoxic T-lymphocytes (CTL) activity and interferon (IFN)-gamma secreting T cells compared with CEA and CpG-ODN (CEA + CpG) or TAT-CEA vaccination alone. Vaccination with TAT-CEA + CpG elicited Th1-based responses, as indicated by the higher ratio of immunoglobulin (Ig)G2a antibody/IgG1 antibodies specific for CEA. The survival rate was significantly increased after vaccination with TAT-CEA + CpG in a tumor model using MC38/CEA2. Furthermore, the TAT-CEA +/- CpG vaccine groups showed similar antitumor immunity to the CEA peptide-pulsed DC (CEA peptide/DC) vaccine groups. These data suggest that coadministration of TAT fusion protein with CpG-ODN may serve as a potential formulation for enhancing antitumor activity.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Apoptosis Regulatory Proteins/therapeutic use , Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Oligodeoxyribonucleotides/therapeutic use , Recombinant Fusion Proteins/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/therapeutic use , Adjuvants, Immunologic/administration & dosage , Animals , Apoptosis Regulatory Proteins/administration & dosage , Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/administration & dosage , Carcinoembryonic Antigen/therapeutic use , Female , Immunotherapy, Adoptive , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Survivin , T-Lymphocytes, Cytotoxic/immunology , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...