Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Eur Rev Med Pharmacol Sci ; 26(15): 5380-5392, 2022 08.
Article in English | MEDLINE | ID: mdl-35993632

ABSTRACT

OBJECTIVE: Poncirus trifoliata (P. trifoliata) fruits exert phytotherapeutic effects, depending on their maturity level. However, the mechanism by which these phytotherapeutic effects are exerted remains undefined - especially in cancers. Therefore, in this study, we investigated the effects of the immature fruit extract of P. trifoliata on a B16 melanoma cell line. MATERIALS AND METHODS: The effect of immature P. trifoliata extract on B16 cells was evaluated by MTT assay, cell proliferation, FACScan analysis of cell cycles, confocal imaging analysis, nuclear (Hoechst) staining, apoptosis assay (Annexin V-fluorescein isothiocyanate/propidium iodide staining), and Western blot assay. The capacity of immature P. trifoliata extract to inhibit the invasion and migration of B16 cells was assessed using the scratch-wound assay and Matrigel migration assay. The effect of immature P. trifoliata extract on mitochondrial function was determined via the mitochondrial membrane potential assay, activity, and fraction and cytosol proteins. RESULTS: Treating B16 cells with a methanol extract of immature P. trifoliata (MEPT) significantly inhibited cell viability, migration, and invasiveness in a dose- (p<0.01) and time (p<0.01)- dependent manner. MEPT arrested the cells in the G1 phase of the cell cycle and led to the activation of the PI3K/AKT/p21 pathway. Furthermore, MEPT dose-dependently induced apoptosis in B16 cells by increasing the expression of the pro-apoptotic proteins Bax and Apaf-1, while decreasing the expression of the anti-apoptotic protein, Bcl-2. MEPT treatment also decreased mitochondrial membrane potential. CONCLUSIONS: Immature P. trifoliata extract inhibited the growth of melanoma cells by inducing cell apoptosis through mitochondrial pathways. Therefore, further research into immature P. trifoliata extract as a potential therapeutic compound for melanoma treatment is warranted.


Subject(s)
Melanoma , Poncirus , Apoptosis , Cell Line, Tumor , Cell Proliferation , Fruit , Humans , Melanoma/metabolism , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Poncirus/metabolism
2.
J Periodontal Res ; 53(5): 816-824, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29851069

ABSTRACT

BACKGROUND AND OBJECTIVE: Phelligridin D is a hispidin analogue from the mushroom Phellinus baumii that is widely used as a food source in East Asia. This study tested phelligridin D for the anti-inflammatory effect and mechanism in lipopolysaccharide (LPS)-induced human periodontal ligament cells (HPDLCs). The objective of this study was to clarify whether the anti-inflammatory function of phelligridin D affects periodontal regeneration for supporting the HPDLCs of teeth. MATERIAL AND METHODS: Primary HPDLCs were isolated from healthy teeth and then cultured. The anti-inflammatory function, mechanism and differentiation molecules were verified with reactive oxygen species generation and western blot analysis in LPS-induced HPDLCs. RESULTS: HPDLCs showed increased inflammatory molecules (intracellular adhesion molecule-1 and vascular cell adhesion molecule-1) and decreased osteogenic proteins (bone morphogenetic protein-7, Osterix and runt-related transcription factor 2) by LPS treatment. Phelligridin D decreased inflammatory molecules and increased osteogenic molecules via downregulation of the extracellular signal-regulated kinase and c-jun N-terminal kinases pathway among the mitogen-activated protein kinase, followed by blocking of nuclear factor kappa-B translocation from cytosol to nucleus. In addition, phelligridin D showed antioxidant properties by reducing reactive oxygen species activity. Finally, the anti-inflammatory and antioxidant function of phelligridin D promoted the periodontal differentiation of HPDLCs. CONCLUSION: These results suggest that phelligridin D supports teeth on the alveolar bone against outside stress, and may be used as an anti-inflammatory compound for the prevention of periodontitis or periodontal regenerative related disease.


Subject(s)
Anti-Inflammatory Agents , Cell Differentiation/drug effects , Lipopolysaccharides/adverse effects , Periodontal Ligament/drug effects , Periodontal Ligament/physiology , Pyrones/pharmacology , Regeneration/drug effects , Agaricales/chemistry , Bone Morphogenetic Protein 7/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Periodontitis/prevention & control , Pyrones/isolation & purification , Sp7 Transcription Factor/metabolism , Stimulation, Chemical , Vascular Cell Adhesion Molecule-1/metabolism
3.
Int Endod J ; 51(4): 438-447, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28898431

ABSTRACT

AIM: To examine the properties of Schisandrin C as an anti-inflammatory and antioxidant compound, and whether its characteristics promote mitochondrial biogenesis in human dental pulp cells (HDPCs). METHODOLOGY: HDPCs were extracted from fresh third molars and cultured for experiments. Reactive oxidative stress (ROS) and nitric oxide (NO) formation were analysed by a Muse cell analyser. Western blotting and gelatin zymography were used to identify the presence of antioxidants, as well as anti-inflammatory and mitochondrial biogenesis with specific antibody. An unpaired Student's t-test was used for statistical analysis. RESULTS: Schisandrin C inhibited lipopolysaccharide-stimulated inflammatory molecules; interleukin 1 beta, tumour necrosis factor alpha, intracellular adhesion molecule-1, vascular cell adhesion molecule-1, matrix metalloproteinase-2 and -9, NO production, ROS formation, nuclear factor kappa B translocation (P < 0.05) through the mitogen-activated protein kinase pathway. Schisandrin C increased the expression of superoxide dismutase enzymes as well as haem oxygenase-1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha through the phosphorylated-protein kinase B (p-Akt) and nuclear factor erythroid 2-related factor-2 pathways (P < 0.05). The anti-inflammatory and antioxidant properties of Schisandrin C promoted mitochondrial biogenesis. CONCLUSIONS: Schisandrin C has the potential to reduce inflammation and oxidation and to promote mitochondrial biogenesis. Therefore, Schisandrin C may be considered for use as an anti-inflammatory compound for oral inflammation through mitochondrial biogenesis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Dental Pulp/cytology , Dental Pulp/drug effects , Lignans/pharmacology , Organelle Biogenesis , Polycyclic Compounds/pharmacology , Cyclooctanes/pharmacology , Gelatin , Heme Oxygenase (Decyclizing)/metabolism , Humans , Inflammation/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/adverse effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
J Dent Res ; 95(4): 430-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26661713

ABSTRACT

Diabetes mellitus is closely related to oral-complicated diseases by oxidative stress. This study investigates whether cellular myeloblastosis (c-myb) could protect human dental pulp cells against glucose oxidative stress and regulate autophagy activity for pulp vitality. Diabetes mellitus was induced by streptozotocin in Sprague-Dawley rats, and their pulp tissue in teeth was analyzed in terms of pulp cavity and molecules by hematoxylin and eosin and immunohistochemistry staining. Human dental pulp cells were serially subcultured and treated with glucose oxidase in the presence of elevated glucose to generate glucose oxidative stress. The replication-deficient adenovirus c-myb and small interfering RNA c-myb were introduced for c-myb expression. The pulp tissue from the diabetic rats was structurally different from normal tissue in terms of narrow pulp capacity, reduced c-myb, and dentinogenesis molecules. Glucose oxidase treatment decreased c-myb and dentinogenesis molecules (bone morphogenetic protein 2 and 7, dentin matrix protein 1, and dentin sialophosphoprotein) in human dental pulp cells. However, overexpression of c-myb by adenovirus c-myb increased dentinogenesis, autophagy molecules (autophagy protein 5, microtubule-associated protein 1A/1B-light chain 3, and Beclin-1), and cell survival via p-AMPK/AKT signaling even with glucose oxidative stress. In contrast, the lack of c-myb decreased the above molecules and cell survival by downregulating p-AMPK/AKT signaling. The results indicate that diabetes leads to irreversible damage to dental pulp, which is related to downexpression of autophagy via the p-AMPK/AKT pathway by decline of c-myb. The findings of this study provide a new insight that c-myb could ameliorate autophagy activity and that it is applicable for monitoring complicated diseases of dental pulp. The involvement of c-myb in pulp pathology could serve a therapeutic target in oral-complicated diseases.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/physiology , Dental Pulp/cytology , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-myb/physiology , Animals , Blotting, Western , Cells, Cultured , Dentinogenesis/drug effects , Glucose Oxidase/pharmacology , Humans , Immunohistochemistry , Male , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Transfection
5.
J Dent Res ; 94(5): 729-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25744070

ABSTRACT

This study investigated the relevance between pulp vitality and autophagy in aged human dental pulp cells (HDPCs) and whether peroxisome proliferator-activated receptor gamma (PPARγ) affects autophagy regulation for homeostasis in the aging progress. In vivo experiments were used in human and Sprague-Dawley rat teeth obtained from young and adult individuals. Aging- and autophagy-related molecules were determined by immunohistochemistry and hematoxylin and eosin staining. HDPCs were serially subcultured until spontaneously arrested for in vitro aging, and the replication deficiency adenovirus was introduced for PPARγ overexpression. Subsequently, the effect of PPARγ on regulation of autophagy molecules, mitochondria activity, and cell viability was assessed using Western blotting, confocal microscopy, and the MTT assay, respectively. In adult pulp tissue, autophagy molecules (autophagy protein 5, microtubule-associated protein 1A/1B light chain, and Beclin-1) were increased, but aging-related (PPARγ and heme oxygenase 1 [HO-1]) and dentinogenesis (dentin sialophosphoprotein and dentin matrix acidic phosphoprotein) molecules were decreased. In aged HDPCs, autophagy and intercellular adhesion molecule 1 and vascular cell adhesion molecule 1 were increased, while PPARγ and HO-1 were decreased. Under stimulation with lipopolysaccharide, autophagy- and aging-related molecules were differentially expressed between young and aged cells. PPARγ induced HO-1 and autophagy molecules but reduced inflammatory molecules in aged cells. In addition, PPARγ activated strong mitochondrial activity and cell viability in aging cells. Inhibition of HO-1 by tin protoporphyrin IX exacerbated autophagy and mitochondrial activity as well as cell viability in young cells. This study indicates that PPARγ maintains pulp homeostasis through the regulation of autophagy molecules during the life span of HDPCs.


Subject(s)
Aging/physiology , Autophagy/physiology , Dental Pulp/cytology , Homeostasis/physiology , PPAR gamma/physiology , Adult , Aged , Animals , Apoptosis Regulatory Proteins/analysis , Autophagy/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/analysis , Heme Oxygenase-1/analysis , Heme Oxygenase-1/antagonists & inhibitors , Humans , Intercellular Adhesion Molecule-1/analysis , Lipopolysaccharides/pharmacology , Metalloporphyrins/pharmacology , Microtubule-Associated Proteins/analysis , Middle Aged , Mitochondria/drug effects , Mitochondria/physiology , PPAR gamma/analysis , Phosphoproteins/analysis , Protoporphyrins/pharmacology , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/analysis , Vascular Cell Adhesion Molecule-1/analysis , Young Adult
6.
Braz. j. med. biol. res ; 48(3): 226-233, 03/2015. graf
Article in English | LILACS | ID: lil-741251

ABSTRACT

Peroxisome proliferator activator receptor-gamma (PPARγ) is a ligand-activated transcriptional factor involved in the carcinogenesis of various cancers. Insulin-like growth factor-binding protein-3 (IGFBP-3) is a tumor suppressor gene that has anti-apoptotic activity. The purpose of this study was to investigate the anticancer mechanism of PPARγ with respect to IGFBP-3. PPARγ was overexpressed in SNU-668 gastric cancer cells using an adenovirus gene transfer system. The cells in which PPARγ was overexpressed exhibited growth inhibition, induction of apoptosis, and a significant increase in IGFBP-3 expression. We investigated the underlying molecular mechanisms of PPARγ in SNU-668 cells using an IGFBP-3 promoter/luciferase reporter system. Luciferase activity was increased up to 15-fold in PPARγ transfected cells, suggesting that PPARγ may directly interact with IGFBP-3 promoter to induce its expression. Deletion analysis of the IGFBP-3 promoter showed that luciferase activity was markedly reduced in cells without putative p53-binding sites (-Δ1755, -Δ1795). This suggests that the critical PPARγ-response region is located within the p53-binding region of the IGFBP-3 promoter. We further demonstrated an increase in PPARγ-induced luciferase activity even in cells treated with siRNA to silence p53 expression. Taken together, these data suggest that PPARγ exhibits its anticancer effect by increasing IGFBP-3 expression, and that IGFBP-3 is a significant tumor suppressor.


Subject(s)
Adult , Female , Humans , Male , Asthma/chemically induced , Genes, MHC Class I/genetics , Genes, MHC Class II/genetics , Isocyanates/toxicity , Occupational Diseases/chemically induced , Occupational Exposure/adverse effects , Asthma/genetics , Genetic Variation , Genotype , Occupational Diseases/genetics , Polymorphism, Single Nucleotide , Risk
7.
Braz J Med Biol Res ; 48(3): 226-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25590353

ABSTRACT

Peroxisome proliferator activator receptor-gamma (PPARγ) is a ligand-activated transcriptional factor involved in the carcinogenesis of various cancers. Insulin-like growth factor-binding protein-3 (IGFBP-3) is a tumor suppressor gene that has anti-apoptotic activity. The purpose of this study was to investigate the anticancer mechanism of PPARγ with respect to IGFBP-3. PPARγ was overexpressed in SNU-668 gastric cancer cells using an adenovirus gene transfer system. The cells in which PPARγ was overexpressed exhibited growth inhibition, induction of apoptosis, and a significant increase in IGFBP-3 expression. We investigated the underlying molecular mechanisms of PPARγ in SNU-668 cells using an IGFBP-3 promoter/luciferase reporter system. Luciferase activity was increased up to 15-fold in PPARγ transfected cells, suggesting that PPARγ may directly interact with IGFBP-3 promoter to induce its expression. Deletion analysis of the IGFBP-3 promoter showed that luciferase activity was markedly reduced in cells without putative p53-binding sites (-Δ1755, -Δ1795). This suggests that the critical PPARγ-response region is located within the p53-binding region of the IGFBP-3 promoter. We further demonstrated an increase in PPARγ-induced luciferase activity even in cells treated with siRNA to silence p53 expression. Taken together, these data suggest that PPARγ exhibits its anticancer effect by increasing IGFBP-3 expression, and that IGFBP-3 is a significant tumor suppressor.


Subject(s)
Apoptosis/drug effects , Insulin-Like Growth Factor Binding Protein 3/metabolism , PPAR gamma/metabolism , Stomach Neoplasms/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , PPAR gamma/genetics , Signal Transduction , Stomach Neoplasms/metabolism , Transcriptional Activation , Up-Regulation
8.
Biol Sport ; 31(1): 73-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-25187675

ABSTRACT

The study investigated the effect of high- and low-intensity exercise training on inflammatory reaction of blood and skeletal muscle in streptozotocin (STZ)-induced diabetic male Sprague-Dawley rats (243 ± 7 g, 8 weeks). The rats completed treadmill running in either high-intensity exercise (6 weeks of exercise training, acute bouts of exercise) or low-intensity exercise (6 weeks of exercise training). Non-running, sedentary rats served as controls. To induce diabetes mellitus, rats received a peritoneal injection of STZ (50 mg · kg(-1)). Rats were sacrificed immediately after an acute bout of exercise and 6 weeks of exercise training. Inflammatory factors were analyzed by ELISA and by immune blotting from the soleus and extensor digitorum longus muscles. In the serum, inflammatory cytokines (IL-1ß, TNF-α, IL-6, IL-4) and reactive oxygen species (ROS) (nitric oxide and malondialdehyde) increased in diabetic rats. However, all exercise training groups displayed reduced inflammatory cytokines and reactive oxygen species. In skeletal muscles, low-intensity exercise training, but not high intensity exercise, reduced the levels of COX-2, iNOS, and MMP-2, which were otherwise markedly elevated in the presence of STZ. Moreover, the levels of GLUT-4 and MyoD were effectively increased by different exercise intensity and exercise duration. Low-intensity exercise training appeared most effective to reduce diabetes-related inflammation. However, high-intensity training also reduced inflammatory factors in tissue-specific muscles. The data implicate regular exercise in protecting against chronic inflammatory diseases, such as diabetes.

9.
Oral Dis ; 20(8): 827-32, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24924779

ABSTRACT

OBJECTIVES: The aim of this study was to elucidate the role of 6-6 bieckol (EB1) and pholorofucofuroeckol-A (EB5) from brown seaweed marine algae (Eisenia bicyclis) on lipopolysaccharide (LPS)-induced inflammation in human dental pulp cells (HDPCs). METHODS: The cytotoxicity of EB1 and EB5 was examined by MTT assay on LPS-induced human dental pulp cells. Their role on expression of inflammatory, odontogenic, and osteogenic molecules was determined by Western blot analysis. The dentin mineralization was checked by alkaline phosphatase activity. RESULTS: The five compounds from E. bicyclis have different structure with non-cytotoxic in HDPCs. EB1 and EB5 showed anti-inflammatory properties and inhibited phosphorylated-extracellular signal-regulated kinase (p-ERK1/2) and phosphorylated-c-jun N-terminal kinases (p-JNK) without any cytotoxicity. In particular, EB1 inhibited cyclooxygenase-2 (COX-2) and p-ERK1/2 signaling, and EB5 inhibited only p-ERK1/2 signaling but not COX-2. Both compounds inhibited nuclear factor kappa-B (NF-κB) translocation. Furthermore, EB1 and EB5 increased dentinogenic and osteogenic molecules, and dentin mineralized via alkaline phosphatase activity (ALP) in LPS-induced HDPCs. CONCLUSIONS: This study elucidates that EB1 and EB5 have different types of anti-inflammatory property and help in dentin formation. Therefore, these compounds derived from marine algae of E. bicyclis may be used as selective therapeutic strategies for pulpitis and oral diseases.


Subject(s)
Dental Pulp/pathology , Inflammation/pathology , MAP Kinase Signaling System , Cells, Cultured , Cyclooxygenase 2 Inhibitors/pharmacology , Dental Pulp/enzymology , Humans , Inflammation/enzymology , Seaweed
10.
J Dent Res ; 92(9): 840-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23838059

ABSTRACT

Bone regeneration around titanium (Ti) implants is a relatively slow process. The c-myb transcription factor has been associated with high proliferation and differentiation rates in bone. This study analyzed whether c-myb can enhance new bone surrounding the implant. In vitro overexpressed chitosan-gold nanoparticles conjugated with plasmid DNA/c-myb (Ch-GNPs/c-myb)-coated Ti surfaces were associated with enhanced expression of the osteogenic molecules osteopontin (OPN), runt-related transcription factor 2 (RUNX-2), and bone morphogenetic proteins (BMP2/7) in MC-3T3E1 osteoblast cells. Further, to determine its in vivo effect, we inserted Ch-GNPs/c-myb-coated Ti implants into rat mandibles. One and 4 wks post-implantation, mandibles were examined by microcomputed tomography, immunohistochemistry, and hematoxylin & eosin staining. The microcomputed tomography analysis demonstrated that c-myb overexpression increased the density and volume of newly formed bone surrounding the implants, compared with those in controls (p < .05). Further, c-myb increased the number of cells expressing BMP2/7 and aided in the increase of new bone (p < .05). These results support the view that c-myb overexpression accelerates new bone surrounding implants and can serve as a potent molecule in promoting tissue regeneration around dental implants. The recipient rat used in this system provides an excellent in vivo model for studies of bone regeneration.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Dental Implants , Dental Materials/chemistry , Mandible/drug effects , Osteogenesis/drug effects , Proto-Oncogene Proteins c-myb/pharmacology , 3T3 Cells , Animals , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 7/drug effects , Bone Regeneration/drug effects , Chitosan/chemistry , Core Binding Factor Alpha 1 Subunit/drug effects , Gene Transfer Techniques , Gold/chemistry , Male , Mandible/pathology , Mandible/surgery , Mice , Nanoparticles/chemistry , Osteoblasts/drug effects , Osteopontin/drug effects , Rats , Rats, Sprague-Dawley , Titanium/chemistry , Tooth Socket/drug effects , Tooth Socket/pathology , Tooth Socket/surgery
11.
Oral Dis ; 19(2): 193-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22849812

ABSTRACT

OBJECTIVES: Heme oxygenase-1 (HO-1) is contributed to odontoblast differentiation in human dental pulp cells (HDPCs). In this study, pachymic acid from mushroom Formitopsis niagra is examined to determine whether it affects pulpal inflammation and promotes odontogenesis via HO-1 gene expression. MATERIALS AND METHODS: The HDPCs were given H2O2 for inflammation. The anti-inflammatory character and odontoblast differentiation by pachymic acid were analyzed by Western blotting, alkaline phosphatase activity, and alizarin red S staining. To understand the mechanism of pachymic acid via HO-1 induction, the cells were treated with zinc protoporphyrin IX (ZnPP: HO-1 inhibitor). RESULTS: H2O2 induced pulp inflammation and disturbed odontoblast differentiation. However, the HDPCs treated with pachymic acid affected anti-inflammatory effect and induction of odontoblast differentiation through increasing HO-1 expression. In addition, pachymic acid has potent cytoprotection and mineralization under H2O2 treatment. Furthermore, pachymic acid significantly suppressed nuclear factor-kappa B (NF-κB) translocation into nucleus and induced NE-E2-related factor-2 (Nrf2) translocation into nucleus. Overall, NF-κB and Nrf2 translocation were regulated by the HO-1 pathway. CONCLUSIONS: The pachymic acid showed anti-inflammatory function and odontoblast differentiation via HO-1 pathway. These results suggested that pachymic acid may be applicable for prevention of oral inflammation or to improve dentin mineralization against several stresses.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Differentiation/drug effects , Dental Pulp/cytology , Heme Oxygenase-1/physiology , Odontoblasts/cytology , Triterpenes/pharmacology , Cells, Cultured , Humans
12.
Gene Ther ; 14(6): 476-83, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17122804

ABSTRACT

We describe the development of an aerosol system for topical gene delivery to the lungs of C57BL/6 mice. This system is based on the combination of the commercial cationic lipid Lipofectin with a novel amphiphilic triblock copolymer, poly(p-dioxanone-co-L-lactide)-block-poly(ethylene glycol) (PPDO/PLLA-b-PEG, and abbreviated in the text as polymeric micelles). After optimizing conditions for DNA delivery to the lungs of mice using the combination of polymeric micelles with Lipofectin and LacZ DNA, we used the Lipofectin/polymeric micelle system to deliver the tumor suppressor gene PTEN to the lungs of C57BL/6 mice bearing the B16-F10 melanoma. Lipofectin/PTEN/polymeric micelles significantly improved gene expression of PTEN in the lungs of mice with no evidence of cell toxicity or acute inflammation. Importantly, lung metastasis, as measured by lung weight, was significantly reduced (P<0.001), as were total tumor foci in the lungs (P<0.001) and size of individual tumor nodules in animals treated with Lipofectin/PTEN/polymeric micelles compared with control animals. Survival time was also extended. These results suggest that the Lipofectin/polymeric micelle system is appropriate for enhancing gene delivery in vivo and that it can be applied as a non-invasive gene therapy for lung cancer.


Subject(s)
Genetic Therapy/methods , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , PTEN Phosphohydrolase/genetics , Polyesters , Polyethylene Glycols , Aerosols , Animals , Female , Gene Expression , Immunohistochemistry , Lung Neoplasms/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Micelles , PTEN Phosphohydrolase/analysis , PTEN Phosphohydrolase/metabolism , Phosphatidylethanolamines , Transfection/methods , Transgenes
13.
Acta Physiol Scand ; 183(4): 389-97, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15799775

ABSTRACT

AIM: The effects of cold exposure on gene and protein expression of vascular endothelial growth factor (VEGF), in heart and skeletal muscles, were studied in male adult Wistar rats. METHODS: Cold immersion was accomplished by submerging the rats in shoulder-deep water maintained at approximately 18 degrees C, either acutely (1 h) or chronically (1 h day(-1), 5 days week(-1) for 20 weeks). The expressions of VEGF mRNA and protein in heart, gastrocnemius, and soleus muscles were examined by Northern and Western blotting and competitive-polymerase chain reaction techniques. RESULTS: The expressions of VEGF mRNA and protein were markedly increased in cardiac muscle of the cold-immersed group, particularly in the 1-hour exposure group, whereas VEGF mRNA and protein in gastrocnemius were decreased significantly after an acute exposure. Although the protein level in gastrocnemius remained low in the chronically exposed group, the expression of mRNA of VEGF(165) with chronic exposure in this group returned to the control level and that of VEGF(206) was 15% greater than that in controls. The expression of mRNA for VEGF(165) in soleus was also lowered by acute cold exposure, although that for VEGF(206) was stable. However, VEGF protein was increased by 50%. After 20 weeks, all of these parameters were increased over the levels found in the controls. CONCLUSION: These results suggest that the VEGF gene may be a major regulatory factor in cardiac and skeletal muscle adaptation to the cold environment stimulating angiogenesis and thermogenesis.


Subject(s)
Cold Temperature , Heart/physiopathology , Immersion/physiopathology , Muscle, Skeletal/physiopathology , RNA, Messenger/genetics , Vascular Endothelial Growth Factor A/analysis , Actins/analysis , Animals , Gene Expression Regulation/genetics , Male , Muscle Proteins/analysis , Muscle Proteins/genetics , Myocardium/chemistry , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/genetics , Water
14.
Eur J Cancer ; 37(17): 2257-63, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11677116

ABSTRACT

Insulin-like growth factor (IGF)-I and -II are potent mitogens and postulated to exert autocrine, and paracrine effects on growth regulation in human gastric cancer. Their mitogenic effects are tightly regulated by the IGF binding proteins (IGFBPs). In this study, we evaluated the mRNA expression of IGF-I, IGF-II and the IGFBPs in a panel of human gastric cancer cell lines, and normal and tumour tissue specimens from patients with gastric cancer by reverse transcriptase-polymerase chain reaction (RT-PCR) and competitive PCR. Conditioned media (CM) of the gastric cancer cell lines were studied for the secretion of the IGFBPs by western ligand blot (WLB) and western immunoblot (WIB). IGF-I and IGF-II were expressed in all of the gastric cancer cell lines, and the normal and tumour tissue specimens. Overexpression of the IGFs, in particular, IGF-II, was observed in the tumour tissues. The expression pattern of IGFBPs was heterogeneous among the gastric cancer cell lines. IGFBP-2 was expressed in all of the gastric cancer cell lines, whereas IGFBP-1 was not detected in any cell lines. IGFBP-4 was expressed in the most of cell lines. IGFBP-3, IGFBP-5 and IGFBP-6 were expressed in approximately 50% of cell lines. In addition, exogenous IGF-I and IGF-II stimulated the proliferation of gastric cancer cells, suggesting the existence of a functional IGF system in gastric cancer. Taken together, our data-suggest that the IGF-IGFBP system may play an important role in the initiation, progression and metastasis of gastric cancer. Further studies are needed to understand the exact role of IGFs and IGFBPs in gastric neoplasia.


Subject(s)
Adenocarcinoma/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Somatomedins/metabolism , Stomach Neoplasms/metabolism , Adenocarcinoma/pathology , Blotting, Western , Cell Division/drug effects , Gene Expression , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor II/pharmacology , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Tumor Cells, Cultured
15.
Cancer Lett ; 172(1): 83-91, 2001 Oct 22.
Article in English | MEDLINE | ID: mdl-11595133

ABSTRACT

PTEN/MMAC1/TEP1 (PTEN) is a tumor suppressor gene that is mutated in a variety of advanced and metastatic cancers, strongly suggests that PTEN alteration is possibly involved in the tumor progression and formation of metastases. However, the roles of PTEN in tumor growth and metastasis and its functional mechanisms are not fully understood. We evaluated the tumor suppressor function of PTEN gene on tumor growth and metastasis in vitro and in vivo. Our results of in vitro soft agar assay and in vivo PTEN-expressing tumor cell growth showed that PTEN inhibited the tumorigenicity of B16F10 melanoma cells. Anti-metastatic function of PTEN was also revealed by experimental pulmonary metastatic animal model. For the further insight into the mechanisms underlying the PTEN-mediated inhibition of tumor metastasis, we have examined the role of PTEN on the secretion of matrix metalloproteinases (MMPs), insulin-like growth factors (IGFs) and the expression of secretory and cellular vascular endothelial growth factor (VEGF) proteins that have been described to contribute to the metastasis of tumor. PTEN significantly lowered MMPs and IGFs secretion and also expression of secretory and cellular VEGF proteins. These results suggest that PTEN tumor suppressor protein inhibits tumorigenicity and metastasis through regulation of MMP, IGFs, and VEGF expression.


Subject(s)
Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , Tumor Suppressor Proteins , Animals , Blotting, Northern , Blotting, Western , Cell Division , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Endothelial Growth Factors/biosynthesis , Female , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor II/biosynthesis , Lymphokines/biosynthesis , Matrix Metalloproteinases/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mutation , Neoplasm Metastasis , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/biosynthesis , Radioimmunoassay , Time Factors , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Clin Chem Lab Med ; 39(1): 11-4, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11256792

ABSTRACT

The D polymorphism of angiotensin converting enzyme (ACE) gene has been found to be associated with various diseases, and ACE may also be involved in the pathogenesis of erectile dysfunction. On the other hand, interpretation of the data on the association of DD genotype with various diseases is controversial, due to methodological and technical variations in detection of the polymorphisms. We investigated a possible association between the DD genotype and erectile dysfunction in a Korean population, and compared the frequency of ACEgenotypes using our multiplexed PCR method with those based on the conventional PCR method in a sample of erectile dysfunctional and control subjects. There was significant difference in the distribution of ACE genotypes between the erectile dysfunctional (conventional PCR) and the control subjects (multiplexed PCR) (chi2=7.395, p<0.05), but there was no significant difference in the distribution of the genotypes between both groups (chi2=0.815, p>0.05) when our multiplexed PCR method was used. Therefore our results suggest that especially the conventional PCR method for ACE gene polymorphism may require careful control and may need repeated testing to verify the insertion deletion (ID) heterozygotes, and that a multiplexed PCR method can markedly increase the detection rate of the I allele in ID heterozygotes. No association was found between I/D polymorphism and erectile dysfunctional subjects in the Korean population studied.


Subject(s)
Chemistry, Clinical/methods , Erectile Dysfunction/blood , Erectile Dysfunction/genetics , Peptidyl-Dipeptidase A/blood , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Adult , Aged , Alleles , Case-Control Studies , Electrophoresis, Agar Gel , Gene Deletion , Genotype , Heterozygote , Humans , Korea , Male , Middle Aged , Polymerase Chain Reaction , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...