Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
EMBO J ; 39(6): e102214, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32030804

ABSTRACT

Spinal cord microglia contribute to nerve injury-induced neuropathic pain. We have previously demonstrated that toll-like receptor 2 (TLR2) signaling is critical for nerve injury-induced activation of spinal cord microglia, but the responsible endogenous TLR2 agonist has not been identified. Here, we show that nerve injury-induced upregulation of sialyltransferase St3gal2 in sensory neurons leads to an increase in expression of the sialylated glycosphingolipid, GT1b. GT1b ganglioside is axonally transported to the spinal cord dorsal horn and contributes to characteristics of neuropathic pain such as mechanical and thermal hypersensitivity. Spinal cord GT1b functions as an TLR2 agonist and induces proinflammatory microglia activation and central sensitization. Pharmacological inhibition of GT1b synthesis attenuates nerve injury-induced spinal cord microglia activation and pain hypersensitivity. Thus, the St3gal2-GT1b-TLR2 axis may offer a novel therapeutic target for the treatment of neuropathic pain.


Subject(s)
Gangliosides/metabolism , Neuralgia/therapy , Peripheral Nerve Injuries/therapy , Signal Transduction , Toll-Like Receptor 2/agonists , Animals , Gangliosides/antagonists & inhibitors , Gene Expression Regulation , Inflammation , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neuralgia/etiology , Peripheral Nerve Injuries/etiology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells , Sialyltransferases/genetics , Sialyltransferases/metabolism , Spinal Cord/metabolism , Toll-Like Receptor 2/metabolism
2.
Mol Pain ; 13: 1744806917697006, 2017 01.
Article in English | MEDLINE | ID: mdl-28326946

ABSTRACT

Background Accumulating evidence on the causal role of spinal cord microglia activation in the development of neuropathic pain after peripheral nerve injury suggests that microglial activation inhibitors might be useful analgesics for neuropathic pain. Studies also have shown that polyamidoamine dendrimer may function as a drug delivery vehicle to microglia in the central nervous system. In this regard, we developed polyamidoamine dendrimer-conjugated triamcinolone acetonide, a previously identified microglial activation inhibitor, and tested its analgesic efficacy in a mouse peripheral nerve injury model. Result Polyamidoamine dendrimer was delivered selectively to spinal cord microglia upon intrathecal administration. Dendrimer-conjugated triamcinolone acetonide inhibited lipoteichoic acid-induced proinflammatory gene expression in primary glial cells. In addition, dendrimer-conjugated triamcinolone acetonide administration (intrathecal) inhibited peripheral nerve injury-induced spinal cord microglial activation and the expression of pain-related genes in the spinal cord, including Nox2, IL-1ß, TNF-α, and IL-6. Dendrimer-conjugated triamcinolone acetonide administration right after nerve injury almost completely reversed peripheral nerve injury-induced mechanical allodynia for up to three days. Meanwhile, dendrimer-conjugated triamcinolone acetonide administration 1.5 days post injury significantly attenuated mechanical allodynia. Conclusion Our data demonstrate that dendrimer-conjugated triamcinolone acetonide inhibits spinal cord microglia activation and attenuates neuropathic pain after peripheral nerve injury, which has therapeutic implications for the treatment of neuropathic pain.


Subject(s)
Hyperalgesia/etiology , Microglia/drug effects , Peripheral Nerve Injuries/complications , Spinal Cord/pathology , Triamcinolone Acetonide/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Dendrimers/chemistry , Dendrimers/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Peripheral Nerve Injuries/pathology , Triamcinolone Acetonide/chemistry , Triamcinolone Acetonide/therapeutic use
3.
Biochem Biophys Res Commun ; 477(4): 614-619, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27395340

ABSTRACT

A few decades ago, researchers found emerging evidence showing that a number of sequential events lead to the pathological cascade of Alzheimer's disease (AD) which is caused by the accumulation of amyloid beta (Aß), a physiological peptide, in the brain. Therefore, regulation of Aß represents a crucial treatment approach for AD. Neprilysin (NEP), a membrane metallo-endopeptidase, is a rate-limiting peptidase which is known to degrade the amyloid beta peptide. This study investigated soluble NEP (sNEP) produced by recombinant mammalian cells stably transfected with a non-viral NEP expression vector to demonstrate its protective effect against Aß peptides in neuronal cells in vitro. Stably transfected HEK 293 cells were used to purify the soluble protein. sNEP and Aß peptide co-treated hippocampal cells had a decreased level of Aß peptides shown by an increase in cell viability and decrease in apoptosis measured by the CCK-8 and relative caspase-3 activity ratio assays, respectively. This study shows that stably transfected mammalian cells can produce soluble NEP proteins which could be used to protect against Aß accumulation in AD and subsequently neuronal toxicity. Additionally, approaches using protein therapy for potential targets could change the pathological cascade of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Neprilysin/pharmacology , Amyloid beta-Peptides/toxicity , HEK293 Cells , Humans , Recombinant Proteins/pharmacology
4.
J Vasc Surg ; 61(6): 1575-82.e1, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24581482

ABSTRACT

OBJECTIVE: The local delivery of paclitaxel onto a graft has been reported to prevent neointimal hyperplasia. Because more than half of vascular stenoses occur within 3 cm of the venous anastomosis, this study tested the effectiveness of a paclitaxel coating restricted to both ends of the expanded polytetrafluoroethylene (ePTFE) graft to reduce the amount of drug delivered. METHODS: Both ends of ePTFE grafts were coated with paclitaxel at a dose of 0.58 µg/mm(2); the total amount of paclitaxel per graft was 0.66 mg. Paclitaxel-coated hemodialysis grafts 15 cm in length were surgically implanted between the common carotid artery and external jugular vein in female Landrace pigs. The animals were sacrificed 6 weeks after graft placement. Cross sections of the anastomosis sites were analyzed histomorphometrically to measure the ratio of neointimal hyperplasia to the graft area (H/G ratio) and the percentage of luminal stenosis. The experimental results were compared between grafts coated with paclitaxel at the ends only (n = 8), grafts coated over the entire length (n = 6), and uncoated control grafts (n = 6). RESULTS: The mean ± standard error values of the H/G ratios for the arterial anastomosis were 0.82 ± 0.13 (control), 0.41 ± 0.09 (terminal coating), and 0.21 ± 0.04 (whole coating). The values for the venous anastomosis were 0.82 ± 0.12 (control), 0.39 ± 0.11 (terminal coating), and 0.12 ± 0.03 (whole coating). Compared with the uncoated grafts, neointimal hyperplasia was suppressed effectively in the vascular grafts coated terminally with paclitaxel (artery, P = 050; vein, P < .001). However, the suppressive effect was less than that of grafts coated with paclitaxel over the entire length. The percentages of luminal stenosis showed similar tendency to the H/G ratios. CONCLUSIONS: Despite a reduced amount of the drug, paclitaxel coating applied to both ends of the ePTFE hemodialysis grafts effectively suppressed neointimal hyperplasia at the sites of anastomosis.


Subject(s)
Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible , Graft Occlusion, Vascular/prevention & control , Neointima , Paclitaxel/administration & dosage , Renal Dialysis , Animals , Blood Vessel Prosthesis Implantation/adverse effects , Carotid Artery, Common/pathology , Carotid Artery, Common/surgery , Constriction, Pathologic , Disease Models, Animal , Female , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/pathology , Hyperplasia , Jugular Veins/pathology , Jugular Veins/surgery , Polytetrafluoroethylene , Prosthesis Design , Swine , Time Factors
5.
Cell Mol Neurobiol ; 35(7): 1049-59, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25990227

ABSTRACT

Alzheimer's disease (AD) is an age-related disorder that causes a loss of brain function. Hyperphosphorylation of tau and the subsequent formation of intracellular neurofibrillary tangles (NFTs) are implicated in the pathogenesis of AD. Hyperphosphorylated tau accumulates into insoluble paired helical filaments that aggregate into NFTs; therefore, regulation of tau phosphorylation represents an important treatment approach for AD. Heat shock protein 27 (Hsp27) plays a specific role in human neurodegenerative diseases; however, few studies have examined its therapeutic effect. In this study, we induced tau hyperphosphorylation using okadaic acid, which is a protein phosphatase inhibitor, and generated a fusion protein of Hsp27 and the protein transduction domain of the HIV Tat protein (Tat-Hsp27) to enhance the delivery of Hsp27. We treated Tat-Hsp27 to SH-SY5Y neuroblastoma cells for 2 h; the transduction level was proportional to the Tat-hsp27 concentration. Additionally, Tat-Hsp27 reduced the level of hyperphosphorylated tau and protected cells from apoptotic cell death caused by abnormal tau aggregates. These results reveal that Hsp27 represents a valuable protein therapeutic for AD.


Subject(s)
HSP27 Heat-Shock Proteins/administration & dosage , Neuroblastoma/metabolism , Okadaic Acid/toxicity , Recombinant Fusion Proteins/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tau Proteins/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Heat-Shock Proteins , Humans , Molecular Chaperones , Neuroprotective Agents/administration & dosage , Phosphorylation/drug effects , Phosphorylation/physiology
6.
Mol Ther ; 20(4): 829-39, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22252450

ABSTRACT

Noninvasive intranasal drug administration has been noted to allow direct delivery of drugs to the brain. In the present study, the therapeutic efficacy of intranasal small interfering RNA (siRNA) delivery was investigated in the postischemic rat brain. Fluorescein isothiocyanate (FITC)-labeled control siRNA was delivered intranasally in normal adult rats using e-PAM-R, a biodegradable PAMAM dendrimer, as gene carrier. Florescence-tagged siRNA was found in the cytoplasm and processes of neurons and of glial cells in many brain regions, including the hypothalamus, amygdala, cerebral cortex, and striatum, in 1 hour after infusion, and the FITC-fluorescence was continuously detected for at least 12 hours. When siRNA for high mobility group box 1 (HMGB1), which functions as an endogenous danger molecule and aggravates inflammation, was delivered intranasally, the target gene was significantly depleted in many brain regions, including the prefrontal cortex and striatum. More importantly, intranasal delivery of HMGB1 siRNA markedly suppressed infarct volume in the postischemic rat brain (maximal reduction to 42.8 ± 5.6% at 48 hours after 60 minutes middle cerebral artery occlusion (MCAO)) and this protective effect was manifested by recoveries from neurological and behavioral deficits. These results indicate that the intranasal delivery of HMGB1 siRNA offers an efficient means of gene knockdown-mediated therapy in the ischemic brain.


Subject(s)
Brain Ischemia/prevention & control , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Administration, Intranasal , Animals , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Immunoblotting , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
7.
J Gene Med ; 14(4): 272-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22407991

ABSTRACT

BACKGROUND: The development of an efficient method to improve the wound healing process is urgently required for diabetic patients suffering a threat of limb amputations. Various growth factors have been proposed for treatment; however, more research still has to be carried out to maintain their curative effect. In the present study, we describe a simple nonviral gene therapy method for improving wound healing. METHODS: Minicircle plasmid DNA encoding vascular endothelial growth factor (VEGF) was combined with an arginine-grafted cationic dendrimer, PAM-RG4. The formed complexes were injected subcutaneously into the skin wounds of diabetic mice. RESULTS: Actively proliferating cells in wound tissue were efficiently transfected, resulting in a high level of VEGF expression. Within 6 days after injection, skin wounds in the diabetic mice were generally healed and displayed a well-ordered dermal structure, which was confirmed by histological staining. CONCLUSIONS: This simple and effective gene therapy method may represent a powerful tool for the treatment of diabetic foot ulcers and other diseases that are refractory to treatment.


Subject(s)
DNA, Circular/administration & dosage , Dendrimers/chemistry , Diabetes Complications/therapy , Skin/pathology , Vascular Endothelial Growth Factor A/genetics , Wound Healing , Animals , Arginine/chemistry , Cations , Diabetes Complications/pathology , Genetic Therapy , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Skin/blood supply , Vascular Endothelial Growth Factor A/metabolism
8.
J Vasc Surg ; 55(3): 806-814.e1, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22226184

ABSTRACT

OBJECTIVES: Paclitaxel coating of hemodialysis grafts is effective in suppressing neointimal hyperplasia in the graft and vascular anastomosis sites. However, paclitaxel can have unwanted effects on the surrounding tissues. To reduce such problems, we developed a method to coat the drug only on the luminal surface of the graft, with little loading on the outer surface. METHODS: A peristaltic pump and a double-solvent (water and acetone) system were used to achieve an inner coating of paclitaxel. At the ratio of 90% acetone, paclitaxel was homogeneously coated only on the luminal surface of the graft without changing the physical properties. To determine its effect, grafts were implanted between the common carotid artery and the external jugular vein in pigs using uncoated control grafts (n = 6) and low-dose (n = 6, 0.22 µg/mm(2)) and high-dose (n = 6, 0.69 µg/mm(2)) paclitaxel inner-coated grafts. Cross-sections of graft-venous anastomoses were analyzed histomorphometrically 6 weeks after placement to measure the patency rate, percentage of luminal stenosis, and neointimal area. RESULTS: No signs of infection or bacterial contamination were observed in the paclitaxel inner-coated groups. Only one of the six control grafts was patent, but all of the paclitaxel-coated grafts were patent, with little neointima. The mean ± standard error values of percentage luminal stenosis were 75.7% ± 12.7% (control), 17.5% ± 3.1% (low dose), and 19.7% ± 3.0% (high dose). The values for the neointimal area (in mm(2)) were 8.77 ± 1.66 (control), 3.53 ± 0.73 (lose dose), and 4.24 ± 0.99 (high dose). Compared with the control group, paclitaxel inner-coated vascular grafts significantly suppressed neointimal hyperplasia (low dose, P = .001; high dose, P = .002). Myofibroblast proliferation and migration into the graft interstices confirmed the firm attachment of the implanted graft to the surrounding tissue. CONCLUSIONS: Paclitaxel coating on the inner luminal surface of vascular grafts was effective in suppressing neointimal hyperplasia, with little inhibition of myofibroblast infiltration within the graft wall.


Subject(s)
Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible , Drug Carriers , Graft Occlusion, Vascular/prevention & control , Paclitaxel/administration & dosage , Renal Dialysis , Tunica Intima/drug effects , Animals , Blood Vessel Prosthesis Implantation/adverse effects , Cardiovascular Agents/chemistry , Carotid Artery, Common/drug effects , Carotid Artery, Common/pathology , Carotid Artery, Common/surgery , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/pathology , Hyperplasia , Jugular Veins/drug effects , Jugular Veins/pathology , Jugular Veins/surgery , Myofibroblasts/drug effects , Myofibroblasts/pathology , Paclitaxel/chemistry , Prosthesis Design , Solubility , Swine , Time Factors , Tunica Intima/pathology , Tunica Intima/surgery , Vascular Patency/drug effects
9.
Nephrol Dial Transplant ; 27(5): 1997-2004, 2012 May.
Article in English | MEDLINE | ID: mdl-22058173

ABSTRACT

BACKGROUND: Haemodialysis vascular access dysfunction caused by aggressive venous neointimal hyperplasia is a major problem for haemodialysis patients with synthetic arteriovenous (AV) grafts. Several different strategies to prevent venous stenosis by inhibiting smooth muscle cell proliferation and migration using local delivery of potent antiproliferative agents are currently under investigation. We performed this study to evaluate the efficacy of sirolimus-eluting vascular grafts in preventing stenosis and to compare the effectiveness of sirolimus-coated grafts with that of paclitaxel-coated vascular grafts that we characterized in a previous study. METHODS: AV grafts were implanted laterally between the common carotid artery and external jugular vein of 14 female Landrace pigs. Three types of grafts were implanted: grafts coated with 1.08 µg/mm(2) sirolimus (low dose, n = 6), grafts coated with 2.41 µg/mm(2) sirolimus (high dose, n = 2) and uncoated control grafts (n = 6). Animals were sacrificed 6 weeks after surgery. Cross-sections of the venous anastomoses were analysed to determine the percentage of luminal stenosis in each group, and immunohistochemistry was performed to identify the cellular phenotypes of the neointimal hyperplasia and tissues adjacent to the implanted grafts. RESULTS: Compared with the control group, neointimal hyperplasia in the venous anastomoses of the groups implanted with sirolimus-coated vascular grafts was significantly suppressed without infection. The mean ± standard error values for the percentage of luminal stenosis were 75.7 ± 12.7% in the control group and 22.2 ± 1.41% in the low-dose sirolimus-coated group. Myofibroblasts and fibroblasts were the major cell types found in the neointimal hyperplasia. CONCLUSIONS: Neointimal hyperplasia was effectively suppressed by sirolimus-eluting grafts. However, the inhibitory effects of sirolimus-eluting grafts were weaker than those observed for paclitaxel-coated grafts in our previous study.


Subject(s)
Neointima/pathology , Neointima/prevention & control , Paclitaxel/therapeutic use , Polytetrafluoroethylene/therapeutic use , Renal Dialysis/methods , Sirolimus/therapeutic use , Vascular Grafting/methods , Animals , Arteriovenous Shunt, Surgical/methods , Carotid Arteries/surgery , Cell Proliferation/drug effects , Constriction, Pathologic/pathology , Constriction, Pathologic/prevention & control , Female , Graft Occlusion, Vascular/pathology , Graft Occlusion, Vascular/prevention & control , Hyperplasia/pathology , Hyperplasia/prevention & control , Jugular Veins/surgery , Models, Animal , Sirolimus/pharmacology , Swine
10.
Drug Dev Ind Pharm ; 37(1): 41-6, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20950058

ABSTRACT

BACKGROUND: A specific and effective strategy is in demand to treat ovarian cancer successfully. Epidermal growth factor receptor (EGFR) is highly expressed in ovarian cancer, and thus EGFR antisense gene therapy can be a potential therapeutic strategy. METHOD: L-Arginine-grafted-polyamidoamine dendrimer (PAMAM-Arg) has been reported to be a novel nonviral gene delivery carrier. Therefore, the ability of PAMAM-Arg in transferring a luciferase gene to ovarian carcinoma SK-OV3 cells has been examined, and the cytotoxicity of the cationic polymer has been investigated. In addition, the suppression of cell proliferation has been evaluated by transferring an EGFR antisense gene to SK-OV3 cells using PAMAM-Arg. Polyethyleneimine (PEI) 25K was used as a positive control. RESULTS: As a result, in vitro gene transfection efficiency of PAMAM-Arg was enhanced with increasing transfection time and N/P ratios. PAMAM-Arg transferred the luciferase gene into cells more efficiently than PEI. In addition, PAMAM-Arg was minimally toxic to the cells whereas PEI 25K was highly toxic. The polyplexes formed by the EGFR antisense gene and PAMAM-Arg significantly reduced thymidine incorporation into the cells suggesting the suppression of cancer cell proliferation. CONCLUSION: These results suggest that a PAMAM-Arg/EGFR antisense gene complex can be used as a safe and efficient therapeutic agent for cancer gene therapy.


Subject(s)
Arginine/chemistry , Dendrimers/chemistry , Gene Transfer Techniques , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Arginine/administration & dosage , Cell Proliferation/drug effects , DNA, Antisense/genetics , Dendrimers/administration & dosage , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Genetic Therapy , Humans , Luciferases/biosynthesis , Luciferases/genetics , Models, Molecular , Ovarian Neoplasms/enzymology , Polyethyleneimine/chemistry , Thymidine/chemistry , Tumor Cells, Cultured
11.
Catheter Cardiovasc Interv ; 74(6): 881-8, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19496118

ABSTRACT

OBJECTIVE: To evaluate the efficacy of the curcumin-coating stent (CCS) on the inhibition of restenosis in a rabbit iliac artery stent model. RESULTS: Curcumin, pigment naturally acquired from the rhizome of the plant curcuma longa, is known to have antiproliferative, antimigratory, and anti-inflammatory effects. However, it is still unclear that curcumin can inhibit neointimal proliferation of the injured vessel. METHODS: Dose-dependent inhibition of cell growth was observed over a dose range from 10 nM to 10 microM. CCS was prepared by a dip-coating method (high-dose: HD, low-dose: LD). The release profile of the HD CCS showed that drug release persisted until day 21. Scanning electron microscopy of the CCS showed an intact surface of the stent even after expansion. To test the efficacy of CCS in vivo, LD CCS, HD CCS, and bare metal stents (BMS) were implanted in random order in one iliac artery (N = 30 arteries) of male New Zealand White rabbits (N = 15). RESULTS: After 28 days, the LD and HD CCS groups had a 43% and 55% reduction in the neointimal area, compared with the BMS group (BMS 3.3 +/- 1.0 mm(2), LD 1.9 +/- 0.8 mm(2), and HD 0.9 +/- 0.5 mm(2), P < 0.05). There appeared to be no cytotoxicity related to curcumin at the indicated doses. CONCLUSIONS: Curcumin, a natural compound in the human diet, seems to be a safe and effective candidate drug for use in a drug-eluting stent for the prevention of stent restenosis following angioplasty.


Subject(s)
Angioplasty, Balloon/instrumentation , Arterial Occlusive Diseases/prevention & control , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible , Curcumin/administration & dosage , Drug-Eluting Stents/adverse effects , Hypercholesterolemia/complications , Iliac Artery/drug effects , Angioplasty, Balloon/adverse effects , Animals , Arterial Occlusive Diseases/etiology , Arterial Occlusive Diseases/pathology , Becaplermin , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Constriction, Pathologic , Disease Models, Animal , Dose-Response Relationship, Drug , Iliac Artery/pathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Platelet-Derived Growth Factor/metabolism , Prosthesis Design , Proto-Oncogene Proteins c-sis , Rabbits , Rats , Rats, Sprague-Dawley , Surface Properties , Time Factors
12.
Int J Pharm ; 363(1-2): 199-205, 2008 Nov 03.
Article in English | MEDLINE | ID: mdl-18718514

ABSTRACT

It is a matter of concern to develop and design synthetic non-viral gene carriers with high transfection efficiency and low cytotoxicity in gene therapy. Recently, various arginine conjugated dendrimers showed better performance in transfection and greater viability than polyethyleneimine (PEI). In this study, we synthesized and investigated e-PAM-R G2, 3 and 4 which are biodegradable polyamidoamine (PAMAM) dendrimers modified with arginine and compared that with PAMAM-R series containing amide bonds for gene carriers. For plasmid DNA delivery, the transfection efficiency of e-PAM-R G4 was higher than G3 and G2 and similar to PAMAM-R G4 with favorable cell viability. Moreover, they indicated significantly higher suppression of TEL/AML1 protein, maybe due to rapid olidonucleotide (ODNs) release through biodegradability of e-PAM-R. These results suggest that biodegradable and non-toxic e-PAM-R may be useful carriers for the gene including plasmid DNA, antisense ODNs and si-RNA.


Subject(s)
Arginine/metabolism , Cell Nucleus/metabolism , DNA/metabolism , Polyamines/metabolism , Transfection , Active Transport, Cell Nucleus , Arginine/analogs & derivatives , Arginine/chemical synthesis , Arginine/toxicity , Artificial Gene Fusion , Cell Survival/drug effects , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA/chemistry , DNA, Catalytic/genetics , DNA, Catalytic/metabolism , Dendrimers , Dose-Response Relationship, Drug , Down-Regulation , Humans , Hydrolysis , Magnetic Resonance Spectroscopy , Oligonucleotides, Antisense/metabolism , Polyamines/chemical synthesis , Polyamines/toxicity , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , ETS Translocation Variant 6 Protein
13.
J Neurosci ; 26(24): 6413-21, 2006 Jun 14.
Article in English | MEDLINE | ID: mdl-16775128

ABSTRACT

Cerebral ischemic injury proceeds with excitotoxicity-induced acute neuronal death in the ischemic core and with delayed damage processes in the penumbra. However, knowledge concerning the direct mediators that connect these two processes is limited. Here, we demonstrate that high-mobility group box 1 (HMGB1), a nonhistone DNA-binding protein, is massively released into the extracellular space immediately after ischemic insult and that it subsequently induces neuroinflammation in the postischemic brain. Short hairpin (sh)RNA-mediated HMGB1 downregulation in the postischemic brain suppressed infarct size, microglia activation, and proinflammatory marker induction, indicating that HMGB1 plays a crucial role in the inflammatory process. The proinflammatory cytokine-like function of extracellular HMGB1 was further verified in primary cortical cultures and microglial cultures. HMGB1 was found to accumulate in NMDA-treated primary cortical culture media, and supernatants collected from these cultures were found to trigger microglia activation, the hallmark of brain inflammation. Moreover, treatment with recombinant HMGB1 also induced microglial activation, but HMGB1-depleted supernatant produced by anti-HMGB1 antibody treatment or by HMGB1 shRNA expression did not, thus demonstrating the essential role of HMGB1 in microglial activation. Together, these results indicate that HMGB1 functions as a novel proinflammatory cytokine-like factor that connects excitotoxicity-induced acute damage processes and delayed inflammatory processes in the postischemic brain.


Subject(s)
Brain/metabolism , High Mobility Group Proteins/metabolism , Inflammation/metabolism , Inflammation/pathology , Neurons/metabolism , Repressor Proteins/metabolism , Animals , Blotting, Northern/methods , Blotting, Western/methods , Brain/pathology , Cell Death/physiology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/toxicity , Excitatory Amino Acid Agonists/toxicity , Gene Expression/drug effects , Gene Expression/physiology , HMGB1 Protein , High Mobility Group Proteins/pharmacology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Inflammation/etiology , Male , Mice , Microglia/metabolism , Microglia/physiology , N-Methylaspartate/toxicity , Neurons/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction/methods , Staurosporine/toxicity , Time Factors , Transfection/methods
14.
Biomaterials ; 28(11): 2061-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17196650

ABSTRACT

We synthesized arginine-conjugated polypropylenimine dendrimer G2 (DAB-8), PPI2-R for gene delivery systems. Synthesized PPI2-R could retard plasmid DNA at a weight ratio of 4 completely and PPI2-R polyplexes showed a fluorescence of less than 10% over a charge ratio of 2 by PicoGreen reagent assay, suggesting its good DNA condensing ability. The size of PPI2-R polyplex was measured to about 200nm at a charge ratio of 150. PPI2-R displayed 80-90% cell viability at even a 150microg/mL concentration. Transfection efficiency of PPI2-R was found to be high comparable to that of PEI25kD and to be 8-214 times higher than that of unmodified PPI2 on HeLa and 293 cells. Moreover, PPI2-R showed 4 times higher transfection efficiency than PEI25kD, treating with 10microg pDNA because of its low cytotoxicity on HeLa cells. Finally, PPI2-R showed a transfection efficiency 2-3 times higher than PEI25kD on HUVECs, showing its potency as a gene delivery carrier for primary cells. These results demonstrate that arginine-conjugation of PPI2 is successful in developing a low toxic and highly transfection efficient gene delivery carrier.


Subject(s)
Arginine/pharmacology , Drug Carriers , Drug Delivery Systems/methods , Gene Transfer Techniques , Polyamines/chemistry , Arginine/chemistry , Cell Line , Cell Survival , DNA/chemistry , Dendrimers , Endothelium, Vascular/metabolism , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Models, Chemical , Organic Chemicals/pharmacology , Transfection
15.
Biotechnol Prog ; 23(3): 693-7, 2007.
Article in English | MEDLINE | ID: mdl-17465527

ABSTRACT

The major cause of hemodialysis vascular access dysfunction (HVAD) is the occurrence of stenosis followed by thrombosis at venous anastomosis sites due to the aggressive development of venous neointimal hyperplasia. Local delivery of antiproliferative drugs may be effective in inhibiting hyperplasia without causing systemic side effects. We have previously demonstrated that paclitaxel-coated expanded poly(tetrafluoroethylene) (ePTFE) grafts, by a dipping method, could prevent neointimal hyperplasia and stenosis of arteriovenous (AV) hemodialysis grafts, especially at the graft-venous anastomoses; however, large quntities of initial burst release have remained a problem. To achieve controlled drug release, paclitaxel (Ptx)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (Ptx-PLGA-NPs) were prepared by the emulsion-solvent evaporation method and then transferred to the luminal surface and inner part of ePTFE vascular grafts through our micro tube pumping and spin penetration techniques. Scanning electron microscope (SEM) images of various stages of Ptx-PLGA-NPs unequivocally showed that micro tube pumping followed by spin penetration effectively transferred Ptx-PLGA-NPs to the inner part, as well as the luminal surface, of an ePTFE graft. In addition, the in vitro release profiles of paclitaxel demonstrated that this new system achieved controlled drug delivery with a reduced initial burst release. These results suggest that loading of Ptx-PLGA-NPs to the luminal surface and the inner part of an ePTFE graft is a promising strategy to ultimately inhibit the development of venous neointimal hyperplasia.


Subject(s)
Blood Vessel Prosthesis , Lactic Acid/chemistry , Nanoparticles/chemistry , Paclitaxel/chemistry , Polyglycolic Acid/chemistry , Polymers/chemistry , Polytetrafluoroethylene/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Drug Delivery Systems/methods , Humans , Microscopy, Electron, Scanning , Nanoparticles/ultrastructure , Nanotechnology/methods , Paclitaxel/administration & dosage , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer
16.
Biomaterials ; 27(10): 2292-301, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16313954

ABSTRACT

Over the past years, macromolecular biodegradable polymers have been attracting considerable attention as gene delivery vehicles due to their safety and many potential applications. In the process of developing such biopolymers, we synthesized a biodegradable, network-type poly(amino ester) polymer (nt-PAE), which showed desirable chemical properties and a mechanical durability with high transfection efficiency. A small library-like approach to polymer synthesis using melt polycondensation followed by surface modification with aminohexanoic acid resulted in the final polymer, nt-PAE. The network-type structure of nt-PAE consists of biodegradable ester linkages and tertiary amines embedded in the backbone, and primary amines positioned at the surface. Unlike other rapidly degrading ester polymers, the nt-PAE exhibited fairly slow degradation, as the polymer sustained its DNA complexing ability for 10 days under physiological buffer conditions. In addition, the transfection efficiency of the nt-PAE could be increased to the same level comparable with that of PEI even in the presence of serum. Low toxicity and high transfection efficiency with a slow degradation profile implies that the nt-PAE can be used as a valuable nonviral gene delivery system.


Subject(s)
Biocompatible Materials/chemical synthesis , Biocompatible Materials/metabolism , Cations , Drug Carriers , Gene Transfer Techniques , Polymers/chemical synthesis , Polymers/metabolism , Biocompatible Materials/chemistry , Cell Line , Humans , Microscopy, Atomic Force , Molecular Structure , Particle Size , Polymers/chemistry , Transfection
17.
J Control Release ; 114(1): 110-7, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16842881

ABSTRACT

PAMAM-Arg is a cationic arginine-grafted polyamidoamine (PAMAM) dendrimer. In the previous study, we reported that PAMAM-Arg facilitates transfection in a range of mammalian cell types. In the present study, we investigated the transfection efficiency of PAMAM-Arg in primary cortical cultures, which are known to be extremely vulnerable to exogenous gene transfection. PAMAM-Arg/DNA complexes showed particularly high transfection efficiencies and low cytotoxicity in primary cortical cells, as compared to other gene carriers such as, native PAMAM, polyethylenimine (BPEI), and Lipofectamine. Efficient transfection was not limited to neurons but extended to all three glial cells, astrocytes, microglia, and oligodendrocytes, present in these primary cortical cultures. The potential use of PAMAM-Arg was demonstrated by efficient gene knock-down by transfecting HMGB1 shRNA-expressing plasmid. The numbers of green fluorescent protein (GFP)-positive and HMGB1-negative cells indicated that PAMAM-Arg/shRNA-expressing plasmid complex suppressed target gene expression in over 40% of cells, which is the highest level achieved to date in primary cortical culture by any gene carrier. Here, we present evidence of the successful delivery and expression of both a reporter gene and of a shRNA-expressing plasmid in primary cortical cells, which demonstrates the potential of PAMAM-Arg for mediating gene delivery to primary neuronal cells.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Neurons/metabolism , Peptides/chemistry , Polyamines/chemistry , Transfection/methods , Animals , Astrocytes/cytology , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dendrimers , Down-Regulation/genetics , Gene Expression/genetics , Green Fluorescent Proteins/genetics , HMGB1 Protein/genetics , Luciferases/genetics , Mice , Neurons/cytology , Peptides/toxicity , Plasmids/genetics , Polyamines/toxicity , RNA Interference , RNA, Double-Stranded/genetics
18.
Int J Pharm ; 320(1-2): 171-8, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16769187

ABSTRACT

Nuclear membrane is one of the main barriers in polymer-mediated intracellular gene delivery. It was previously reported that glucocorticoid receptor dilated the nuclear pore and translocated into nucleus when it bound to its ligand, glucocorticoid. This suggests that the transport of DNA into nucleus may be facilitated by glucocorticoid. In this study, a glucocorticoid, dexamethasone, was conjugated to polyamidoamine (PAMAM) dendrimer and the effect of the conjugation was investigated. The PAMAM-Dexamethasone (PAM-Dexa) was synthesized by the one-step reaction using Traut's reagent. PAM-Dexa/plasmid DNA complex was completely retarded at a 1/1 weight ratio (polymer/DNA) in a gel retardation assay. PAM-Dexa protected DNA from DNase I for more than 60 min. PAM-Dexa/plasmid DNA complex showed the highest transfection efficiency to 293 cells at a 0.8/1 weight ratio. At this ratio, PAM-Dexa had higher transfection efficiency than PAMAM. Especially in the presence of serum during the transfection, the transfection efficiency of PAM-Dexa was higher than that of PAMAM or PEI by one order of magnitude. In addition, more PAM-Dexa/DNA complexes were observed in the nucleus region than PAMAM/DNA from the confocal microscopy studies. These results indicated that the technique with dexamethasone might be useful for the gene delivery using polymeric gene carriers and the development of efficient polymer vectors.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus/metabolism , DNA/metabolism , Dexamethasone/chemistry , Polyamines/chemistry , Transfection , Animals , Cell Line, Tumor , Cell Survival/drug effects , DNA/chemistry , DNA/genetics , Dendrimers , Deoxyribonuclease I , Dexamethasone/toxicity , Electrophoretic Mobility Shift Assay , Humans , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Luciferases/genetics , Mice , Microscopy, Confocal , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Particle Size , Polyamines/toxicity
19.
Water Res ; 40(1): 45-52, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16360189

ABSTRACT

Two membrane bioreactors were operated at aerobic (DO=6.0mg/L) and anoxic (DO<0.3mg/L) conditions for the treatment of synthetic dye wastewater to determine the effect of dissolved oxygen on membrane filterability. The rate of membrane fouling for the anoxic MBR was five times faster than that for the aerobic MBR. Differences in the nature of the biofilm that was formed on the membrane surface as the result of different DO level was the main factor in the different fouling rates. The biofilm structure was characterized using digital image analysis techniques. Biofilm images were obtained using confocal laser scanning microscopy (CLSM) at various operation points. Structural parameters were then computed from these images using an image analysis software (ISA-2). The structural parameters indicated that the anoxic biofilm was thinner than the aerobic biofilm but the anoxic biofilm was spread out on the membrane surface more uniformly and densely, resulting in the higher membrane fouling. Based on the extracellular polymeric substances (EPS) visualization and quantification, it was also found that EPS, key membrane foulants were spread out more uniformly in the anoxic biofilm in spite of lower amount of EPS compared to that in the aerobic biofilm.


Subject(s)
Biofilms/growth & development , Bioreactors , Oxygen , Waste Disposal, Fluid/methods , Filtration , Membranes, Artificial , Microscopy, Confocal , Permeability
20.
Int J Pharm ; 515(1-2): 186-200, 2016 Dec 30.
Article in English | MEDLINE | ID: mdl-27732896

ABSTRACT

Glioblastoma multiform (GBM) is the most frequent and aggressive form of brain tumors in adults. However, the development of more efficient and safe nonviral vector gene therapy represents a promising therapeutic approach, using a tumor-specific killer gene, named apoptin. In this study, we describe the efficacy of non-viral gene delivery vectors, the amino acid-conjugated PAMAM derivatives (PAMAM-H-R and PAMAM-H-K) in delivering a therapeutic gene, displaying affinity toward human primary glioma cells (GBL-14 cells) and dermal fibroblasts. We analyzed transfection efficiency, using luciferase (Luci) and a pDNA encoding for enhanced fluorescent protein (EGFP), and cytotoxicity in both cells. The results show that transfection efficiency of PAMAM-H-R improved compared to native PAMAM dendrimer, but cytotoxicity of PAMAM-H-R and PAMAM-H-K were very low. We treated both cells with a polyplex formation of PAMAM-H-R or PAMAM-H-K/apoptin, and analyzed their cellular uptake and localization by flow cytometry and confocal microscopy. Furthermore, we analyzed the endosomal escape effect using TEM images, and found that PAMAM-H-R showed very fast escape from endosome to the cytosol. Caspase 3 activity assay, cell cycle distribution, and JC-1 analysis showed apoptosis induced by apoptin in GBL-14 cells. This indicates that PAMAM-H-R can be a potential nonviral vector gene delivery carrier for brain tumor therapy. The present study demonstrates that PAMAM-H-R/apoptin gene polyplex can be used as an effective therapeutic candidate for GBM due to its selective induction of apoptosis in primary glioma cells as a potential nonviral gene delivery carrier for brain tumor therapy.


Subject(s)
Apoptosis/drug effects , Dendrimers/administration & dosage , Dipeptides/administration & dosage , Glioma/drug therapy , Polyamines/administration & dosage , Cell Line, Tumor , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Luciferases/administration & dosage , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL