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










Publication year range
1.
Toxicol Res ; 39(1): 157-167, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36726833

ABSTRACT

Titanium nitride (TiN) is a ceramic material with physical properties such as extreme hardness, high decomposition temperature, defect structure, and gold-yellow color. TiN is generally considered non-toxic and safe; however, hazards have not been identified, especially in workers after inhalation exposure. Here, we conducted a four-week inhalation toxicity study of TiN using a nose-only inhalation exposure system in Sprague-Dawley rats. Rats were exposed to TiN for 4 weeks (6 h a day, 5 days per week) at target concentrations of 45, 90, and 180 mg/m3. Clinical signs, mean body weight changes, hematology, blood biochemistry, necropsy, organ weight, bronchoalveolar lavage fluid analysis, and histopathological findings were observed. Analytical concentrations of the low, middle, and high-concentration groups were 45.55 ± 3.18 mg/m3, 90.69 ± 7.30 mg/m3, and 183.87 ± 15.21 mg/m3, respectively. The mass median aerodynamic diameter (MMAD) for the low, middle, and high-concentration groups were 1.44 ± 0.07 µm, 1.47 ± 0.18 µm, and 1.68 ± 0.16 µm, and the geometric standard deviation (GSD) was 2.24 ± 0.03, 2.31 ± 0.16, and 2.43 ± 0.11, respectively. No systemic adverse effects were observed after inhalation exposure to TiN; however, histopathological findings (increased phagocytic macrophages and alveolar/bronchiolar epithelial hyperplasia) and Bronchoalveolar Lavage Fluid (BALF) analysis (elevated lactate dehydrogenase and gamma-glutamyltransferase values) showed adverse effects on the lungs in the middle and high-concentration groups. Based on these results, the no observed adverse effect concentration (NOAEC) is suggested to be 45 mg/m3.

2.
Toxicol Rep ; 8: 1839-1845, 2021.
Article in English | MEDLINE | ID: mdl-34815948

ABSTRACT

1-Propanol is a colorless volatile liquid at room temperature and is an important industrial alcohol. Workers are potentially exposed to it through inhalation during industrial activities, including manufacturing, sampling, filling, and mixing processes, as well as during cleaning, maintenance, and repair. Consequently, further information and/or testing for inhalation-related toxicological data is required to assess occupational risk. In this study, 80 (40 male and 40 female) F344 rats were exposed to 1-propanol vapors for 13 weeks (6 h a day, 5 days per week) at target concentrations of 0, 500, 1,600, and 5200 ppm in a whole-body inhalation chamber system. Clinical signs, mean body weight changes, food consumption, hematology, blood biochemistry, necropsy, organ weight, and histopathological findings were observed. The exposure concentrations in chambers were 501.30 ± 9.54 ppm, 1605.43 ± 66.55 ppm, and 5202.19 ± 102.74 ppm for the low, middle, and high dose groups, respectively. No changes related to 1-propanol were observed, including histopathological findings, except for mean body weight changes. The significant decrease in mean body weight at a high dose was not considered to be an adverse effect. Based on these results, the no observed adverse effect concentration of 1-propanol was estimated to be 5202.19 ppm.

3.
Asian Pac J Allergy Immunol ; 39(1): 15-24, 2021 Mar.
Article in English | MEDLINE | ID: mdl-31175714

ABSTRACT

BACKGROUND: Magnesium deficiency common in obesity is known to promote chronic low-grade inflammation and aggravate asthma symptoms; however, the effects of magnesium supplementation in obese asthmatic patients have not been investigated. OBJECTIVE: To examine the effects of magnesium co-administration with dexamethasone on airway inflammation in obese mice. METHODS: Female C57BL/6 mice were fed a high-fat diet, sensitized with ovalbumin (OVA) to induce allergic reactions, challenged with aerosolized OVA, and administered dexamethasone (3 mg/kg) with or without magnesium. Bronchial inflammation was analyzed based on the presence of inflammatory cells and cytokines in bronchoalveolar lavage fluid, total and OVA-specific IgE in serum, goblet cells ratios, bronchial wall thickness, and expression of α-smooth muscle actin. RESULTS: In obese mice, co-administration of magnesium and dexamethasone decreased IL-13 in bronchoalveolar lavage fluid and total and OVA-specific IgE in serum, and reduced α-smooth muscle actin-positive areas in the bronchi compared with mice treated with dexamethasone alone. However, no differences were observed in dexamethasone-treated normal-weight mice depending on magnesium supplementation. CONCLUSION: These results suggest that magnesium increases immunosuppressive effects of dexamethasone in airway inflammation aggravated by obesity, suggesting that magnesium supplementation may have a potential in alleviating asthma symptoms in obese patients with reduced responses to corticosteroids.


Subject(s)
Adrenal Cortex Hormones/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Asthma/drug therapy , Dexamethasone/administration & dosage , Immunosuppressive Agents/administration & dosage , Magnesium/administration & dosage , Obesity/drug therapy , Animals , Asthma/blood , Asthma/immunology , Asthma/pathology , Bronchi/drug effects , Bronchi/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Count , Cytokines/immunology , Diet, High-Fat , Female , Immunoglobulin E/blood , Inflammation/blood , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Mice, Inbred C57BL , Obesity/blood , Obesity/immunology , Obesity/pathology , Ovalbumin
4.
Toxicol Res ; 36(4): 285-292, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33005587

ABSTRACT

1-Propanol is used as a solvent for waxes, vegetable oils, resins, cellulose esters, and ethers, and is not considered harmful to humans by food and non-occupational exposures. However, workers are potentially exposed to 1-propanol by inhalation when it is used in the workplace. Thus, inhalation toxicity data are needed to assess the hazard of 1-propanol for workers safety. Five male and five female F344 rats were exposed to 1-propanol vapor for 4-weeks (6 h/day, 5 days/week) at concentrations of 0, 100, 400, and 1600 ppm in a whole-body inhalation chamber system. The actual exposure concentrations were 100.11 ± 5.10, 403.19 ± 12.31, and 1598.08 ± 139.58 ppm for the low, middle, and high dose groups, respectively. No clinical signs, significant mean body weight changes, significant changes of hematology or blood biochemistry results, or histopathological abnormalities were seen related to exposure to the test substance. Under the conditions of this study, the no-observed-adverse-effect level of 1-propanol was over 1600 ppm.

5.
J Toxicol ; 2020: 9172569, 2020.
Article in English | MEDLINE | ID: mdl-33029140

ABSTRACT

We evaluated the toxicity of 1-propanol exposure following repeated inhalation over 28- and 90-day periods in male and female B6C3F1 mice to confirm the potential target organs and to determine the no-observable-adverse-effect levels (NOAELs). Five mice of each sex were exposed to 1-propanol at concentrations of 0, 100, 400, or 1600 ppm for 28 days and showed no consequent toxicity. Following this, ten mice of each sex were exposed at concentrations of 0, 500, 1600, or 5200 ppm for 90 days. We observed no effects on food consumption, body weight, organ weight, clinical signs, hematology and biochemistry parameters, or gross or histological features even at the maximum concentration. Therefore, the NOAEL of inhaled 1-propanol was defined as 5200 ppm (12.8 mg/L) for male and female mice under study conditions.

6.
J Appl Toxicol ; 40(7): 965-978, 2020 07.
Article in English | MEDLINE | ID: mdl-32084673

ABSTRACT

An ethanol extract complex of Descurainia sophia seeds and Peucedanum praeruptorum roots, called BP10A, has antitumor potential against colorectal cancer. In the present study, we evaluated the 28-day oral toxicity and the genotoxicity of BP10A. The subacute toxicity test was done through oral administration to mice. ICR mice (n = 10) received daily oral BP10A doses of 0, 500, 1000 and 2000 mg/kg for 28 consecutive days. During administration, general clinical signs, food consumption, organ weights, and hematologic, biochemical and histopathological parameters in male and female mice were assessed. No significant adverse effects up to the highest dose (2000 mg/kg) were found. The genotoxicity was evaluated using a battery of tests, including an in vitro bacterial reverse mutation (Ames) test, an in vivo micronucleus test using bone marrow cells in ICR mice and a chromosomal aberration test using CHL/IU cells. BP10A did not show any genotoxic signs in the Ames (up to 5000 µg/plate), micronucleus (up to 5000 mg/kg) and the chromosomal aberration tests (550-1750 µg/mL). Therefore, BP10A was considered safe based on the subacute toxicity and genotoxicity results, indicating that it is a useful pharmaceutical material with no adverse toxicity.


Subject(s)
Antineoplastic Agents/toxicity , Apiaceae/chemistry , Brassicaceae/chemistry , Chromans/toxicity , Colorectal Neoplasms/drug therapy , DNA Damage/drug effects , Plant Extracts/toxicity , Administration, Oral , Animals , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred ICR , Models, Animal , Plant Extracts/administration & dosage , Plant Roots/chemistry , Seeds/chemistry , Toxicity Tests
7.
Toxicol Rep ; 5: 213-219, 2018.
Article in English | MEDLINE | ID: mdl-29854591

ABSTRACT

The 28-day repeated inhalation study was applied for hazard assessment of 3-methoxybutyl chloroformate (3-MBCF) in Sprague Dawley rats. Groups of five rats per sex were exposed 6 h/day, 5 days per week for 4 weeks to test substance concentration (ranging from 3 to 12 ppm) using a whole-body exposure system. At the terminal sacrifice, following blood collection and gross pathological examination, organ weights were determined and fixed organs were examined. The micronucleus test was performed using bone marrow cells. Exposure of 3-MBCF induced mortality at concentrations above 6 ppm. Decreases in body weight and food intake, hematologic alterations, organ weight changes, and gross and microscopic findings were seen even at the lowest concentrations of 3 ppm. Histopathology revealed principal test substance exposure correlated with lesions in the respiratory tract in both male and female rats above 3 ppm. Groups of male rats exposed above 6 ppm show microscopic lesions in spleens, livers, testes and epididymides; however, the micronucleated polychromatic erythrocytes frequency in bone marrow cells was not changed. Based on histopathology of the respiratory tract and other organs, the no observed adverse effect level (NOAEL) of 3-MBCF in the present study was less than 3 ppm.

8.
BMC Complement Altern Med ; 17(1): 393, 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28793897

ABSTRACT

BACKGROUND: A variety of anticancer chemotherapeutics induce adverse side effects including myelotoxicity. Dried roots of Phragmites communis Trinius, Phragmitis rhizoma, have been clinically used in traditional folk medicine to relieve various symptoms like fever. In this study, we evaluated the protective effect of the aqueous extract of Phragmitis rhizoma (EPR) against docetaxel-induced myelotoxicity in vitro and in vivo. METHODS: The in vitro myelo-protective effect of EPR was evaluated using the colony forming unit (CFU) assay with hematopoietic progenitor cells. The in vivo efficacy of EPR was evaluated in myelosuppressed C57BL/6 male mice which were induced by repeated intraperitoneal injections of 30 mg/kg docetaxel for 3 times. EPR was orally administered for 4 days to docetaxel-induced myelosuppressed C57BL/6 male mice which were induced by intraperitoneal injection of 30 mg/kg docetaxel for 3 times: Group 1 (vehicle control, n = 10), Group 2 (docetaxel plus vehicle, n = 10), Group 3 (docetaxel plus EPR 30 mg/kg, n = 10), Group 4 (docetaxel plus EPR 100 mg/kg, n = 10) and Group 5 (docetaxel plus EPR 300 mg/kg, n = 10). Whole blood counts were measured automatically, and immune organs were histologically examined. Expression of immunomodulatory cytokines was measured by quantitative real-time polymerase chain reaction or enzyme-linked immunosorbent assay. The toxicity of EPR itself was evaluated in normal human cell lines including IMR-90, foreskin fibroblast and human umbilical vein endothelial cells. The hepatotoxicity of EPR was predicted by multi-parametric assays involving cell viability, caspase 3/7 activity, GSH contents and LDH leakage using the HepaRG hepatic cell line. RESULTS: Co-treatment of EPR or its major component, p-hydroxycinnamic acid, increased the numbers of hematopoietic CFU counts in the docetaxel-induced in vitro myelotoxicity assay system. The in vitro protective effect of EPR against docetaxel toxicity was replicated in a myelosuppressed animal model: white blood cells, neutrophils, lymphocytes and red blood cells rebounded; bone marrow niche and structural integrity of the thymus were preserved; and the expression of immune-stimulating cytokines including IL3, IL6, SCF and GM-CSF was enhanced. Furthermore, EPR and p-hydroxycinnamic acid promoted the proliferation of primary splenocytes and thymocytes. In the toxicity assays, no remarkable signs related with toxicity were observed in all tested normal human cells and HepaRG. CONCLUSIONS: EPR has the potential to ameliorate docetaxel-mediated myelotoxicity in both in vitro and in vivo models. However, the identification of the responsible active components and the precise underlying myelo-protective mechanism of EPR need to be elucidated before novel drug development using EPR can precede.


Subject(s)
Antineoplastic Agents/adverse effects , Bone Marrow/drug effects , Coumaric Acids/pharmacology , Hematopoietic Stem Cells/metabolism , Plant Extracts/pharmacology , Poaceae , Taxoids/adverse effects , Animals , Blood Cells , Bone Marrow Cells , Colony-Stimulating Factors/blood , Docetaxel , Enzyme-Linked Immunosorbent Assay , Fibroblasts , Hematopoiesis , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-3/blood , Interleukin-6/blood , Mice, Inbred C57BL , Propionates , Real-Time Polymerase Chain Reaction , Rhizome , Spleen/drug effects , Stem Cell Factor/blood , Thymus Gland/drug effects
9.
BMC Complement Altern Med ; 16(1): 419, 2016 Oct 26.
Article in English | MEDLINE | ID: mdl-27782842

ABSTRACT

BACKGROUND: Oxaliplatin can induce peripheral neuropathy (OXIPN) as an adverse side effect in cancer patients. Until now, no effective preventive or therapeutic drug has been developed; therefore, the dose-limiting factor of OXIPN is still an obstacle in the use of oxaliplatin to treat cancer patients. In the present study, we report for the first time that the aqueous extract of Lithospermi radix (WLR) can attenuate the OXIPN in both in vitro and in vivo neuropathic models. METHODS: The protective effect of WLR on OXIPN was evaluated in vitro by quantifying nerve growth factor (NGF)-stimulated neurite outgrowth in PC12 cells treated with a combination of oxaliplatin and WLR. The neuroprotective potential of WLR was further confirmed by measuring the changes in nociceptive sensitivities to external mechanical stimuli in neuropathic animals induced by oxaliplatin. Histological and immunohistochemical studies were further done to examine the effect of WLR in mouse spinal cords and footpads. RESULTS: Oxaliplatin-induced neurotoxicity in NGF-stimulated PC12 cells. It could reduce the lengths and branching numbers of neuritis in NGF-stimulated PC12 cells. Co-treatment of WLR rescued the differentiated PC12 cells from the neurotoxicity of oxaliplatin. In a chronic OXIPN animal model, administration of oxaliplatin i.p. induced enhanced nociceptive sensitivity to mechanical stimuli (25.0 to 72.5 % of response rate) along with spinal activation of microglias and astrocytes and loss of intraepidermal nerve fibers in footpads, which is remarkably suppressed by oral administration of WLR (67.5 to 35 % of response rate at the end of experiment). Cytotoxicity of oxaliplatin determined in human cancer cells was not affected irrespective of the presence of WLR. CONCLUSIONS: In conclusion, we demonstrated that WLR can attenuate OXIPN in both in vitro and in vivo experimental models, which may be in part attributed to its anti-inflammatory activity in the spinal cord and its neuroprotective potential in the peripheral nerve system without affecting the anti-tumor potential of oxaliplatin. Therefore, WLR could be considered as a good starting material to develop a novel therapeutic agent targeting OXIPN. However, further studies should be done to elucidate the underlying mechanism such as molecular targets and active constituent(s) in WLR with neuroprotective potential.


Subject(s)
Lithospermum/chemistry , Neurotoxicity Syndromes/drug therapy , Organoplatinum Compounds/toxicity , Peripheral Nervous System Diseases , Plant Extracts/pharmacology , Protective Agents/pharmacology , Animals , Cell Line, Tumor , Ganglia, Spinal/drug effects , Humans , Immunohistochemistry , Male , Mice, Inbred C57BL , Nerve Fibers/drug effects , Oxaliplatin , PC12 Cells , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Plant Extracts/chemistry , Protective Agents/chemistry , Rats
10.
Int Immunopharmacol ; 31: 239-47, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26773771

ABSTRACT

meso-Dihydroguaiaretic acid (MDGA), which is a dibenzylbutane lignin isolated from the ethyl acetate fraction of Saururus chinensis, has various biological activities, including anti-oxidative, anti-inflammatory, anti-bacterial, and neuroprotective effects. However, no report has examined the potential anti-asthmatic activity of MDGA. In this study, we evaluated the protective effects of MDGA on asthmatic responses, particularly airway inflammation and mucus hypersecretion in an ovalbumin (OVA)-induced murine model of asthma. Intragastric administration of MDGA significantly lowered the productions of interleukin (IL)-4, IL-5, IL-13, tumor necrosis-α (TNF-α), eotaxin, monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1), and immunoglobulin (Ig)E in bronchoalveolar lavage fluid (BALF), plasma, or lung tissues. Histological studies showed that MDGA inhibited OVA-induced inflammatory cell infiltration and mucus production in the respiratory tract. Moreover, MDGA markedly attenuated the OVA-induced activations of nuclear factor kappa B (NF-κB), extracellular-signal-regulated kinases 1/2 (ERK1/2), and p38 mitogen-activated protein kinase (p38 MAPK). Together, these results suggest that MDGA effectively inhibits airway inflammation and mucus hypersecretion by downregulating the levels of T helper 2 (Th2) cytokines, chemokines, and adhesion molecules, and inhibiting the activations of NF-κB and MAPKs.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Guaiacol/analogs & derivatives , Lignans/therapeutic use , Pneumonia/drug therapy , Saururaceae/immunology , Animals , Cell Movement/drug effects , Chemokine CCL2/metabolism , Cytokines/metabolism , Female , Guaiacol/therapeutic use , Humans , Immunoglobulin E/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Th2 Cells/immunology , Vascular Cell Adhesion Molecule-1/metabolism
11.
World J Hepatol ; 6(2): 98-106, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24575169

ABSTRACT

AIM: To investigate the effect of methylsulfonylmethane (MSM), recently reported to have anti-cancer effects, in liver cancer cells and transgenic mice. METHODS: Three liver cancer cell lines, HepG2, Huh7-Mock and Huh7-H-ras (G12V), were used. Cell growth was measured by Cell Counting Kit-8 and soft agar assay. Western blot analysis was used to detect caspases, poly (ADP-ribose) polymerase (PARP), and B-cell lymphoma 2 (Bcl-2) expressions. For in vivo study, we administered MSM to H-ras (12V) transgenic mice for 3 mo. RESULTS: MSM decreased the growth of HepG2, Huh7-Mock and Huh7-H-ras (G12V) cells in a dose-dependent manner. That was correlated with significantly increased apoptosis and reduced cell numbers in MSM treated cells. Cleaved caspase-8, cleaved caspase-3 and cleaved PARP were remarkably increased in the liver cancer cells treated with 500 mmol/L of MSM; however, Bcl-2 was slightly decreased in 500 mmol/L. Liver tumor development was greatly inhibited in the H-ras (12V) transgenic mice treated with MSM, compared to control, by showing reduced tumor size and number. Cleaved PARP was significantly increased in non-tumor treated with MSM compared to control. CONCLUSION: Liver injury was also significantly attenuated in the mice treated with MSM. Taken together, all the results suggest that MSM has anti-cancer effects through inducing apoptosis in liver cancer.

12.
Toxicol Appl Pharmacol ; 274(3): 402-7, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24370437

ABSTRACT

Di(2-ethylhexyl) phthalate (DEHP) is used as a plasticizer and is widely dispersed in the environment. In this study, we investigated the effects of maternal exposure to DEHP during pregnancy on neonatal asthma susceptibility using a murine model of asthma induced by ovalbumin (OVA). Pregnant BALB/c mice received DEHP from gestation day 13 to lactation day 21. Their offspring were sensitized on postnatal days (PNDs) 9 and 15 by intraperitoneal injection of 0.5µg OVA with 200µg aluminum hydroxide. On PNDs 22, 23 and 24, live pups received an airway challenge of OVA for 30min. Offspring from pregnant mice that received DEHP showed reductions in inflammatory cell count, interleukin (IL)-4, IL-13, and eotaxin in their bronchoalveolar lavage fluid and in total immunoglobulin E and OVA-specific IgE in their plasma compared with offspring from pregnant mice that did not receive DEHP treatment. These results were consistent with histological analysis and immunoblotting. Maternal exposure to DEHP reduces airway inflammation and mucus production in offspring, with a decrease in inducible nitric oxide synthase (iNOS) in the lung tissue. This study suggests that maternal exposure to DEHP during pregnancy reduces asthmatic responses induced by OVA challenge in offspring. These effects were considered to be closely related to the suppression of Th2 immune responses and iNOS expression.


Subject(s)
Asthma/immunology , Diethylhexyl Phthalate/pharmacology , Maternal Exposure , Plasticizers/pharmacology , Prenatal Exposure Delayed Effects/immunology , Animals , Asthma/chemically induced , Bronchoalveolar Lavage Fluid/chemistry , Chemokines, CC/immunology , Disease Susceptibility/immunology , Female , Immunoglobulin E/blood , Inflammation/chemically induced , Inflammation/immunology , Interleukin-13/immunology , Interleukin-4/immunology , Lactation , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Ovalbumin/adverse effects , Pregnancy
13.
Food Chem Toxicol ; 51: 364-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23116642

ABSTRACT

Platycodin D (PD) is the major triterpene saponin in the root of Platycodon grandiflorum. The aim of the present study was to evaluate the protective effects of PD on bile duct ligation (BDL)-induced cholestasis in mice. Mice were allocated to five groups: sham, BDL alone, and BDL with PD treatment at 1, 2, and 4mg/kg. PD was administered to the mice for 28 consecutive days after the BDL operation. PD treatment of BDL-operated mice decreased serum alanine aminotransferase, serum aspartate aminotransferase, and total bilirubin levels by up to 37%, 31%, and 41%, respectively, in comparison with the levels in mice that underwent BDL alone. PD treatment attenuated oxidative stress, as evidenced by an increase in anti-oxidative enzyme levels glutathione and superoxide dismutase together with a decrease in lipid peroxidation and oxidative stress indices levels of malondialdehyde and nitric oxide. Histopathological studies further confirmed the protective effects of PD on cholestasis-induced hepatic injury and liver fibrosis in mice. In addition, nuclear factor-kappa B and inducible nitric oxide synthase levels significantly decreased after PD treatment, as did the levels of hepatocyte apoptosis. Taken together, these results suggest that PD treatment might be beneficial in cholestasis-induced hepatotoxicity.


Subject(s)
Cholestasis/physiopathology , Liver/drug effects , Liver/pathology , Saponins/pharmacology , Triterpenes/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bile Ducts/surgery , Bilirubin/blood , Cholestasis/pathology , Cholestasis/surgery , Disease Models, Animal , Fibrosis/drug therapy , Glutathione/blood , Ligation , Liver/metabolism , Male , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Superoxide Dismutase/metabolism
14.
J Nutr Sci Vitaminol (Tokyo) ; 58(3): 187-94, 2012.
Article in English | MEDLINE | ID: mdl-22878389

ABSTRACT

The present study aims to evaluate the anti-HCV activity of hotwater extract from Platycodon grandiflorum (BC703) with HCV genotype 1b subgenomic replicon system and investigate its hepatoprotective activity on carbon tetrachloride (CCl(4))-induced acute liver damage in mice. BC703 produced significant hepatoprotective effects against CCl(4)-induced acute hepatic injury by decreasing the activities of serum enzymes, nitric oxide and lipid peroxidation. Histopathological studies further substantiated the protective effect of BC703. Furthermore, BC703 inhibited the HCV RNA replication with an EC(50) value and selective index (CC(50)/EC(50)) of 2.82 µg/mL and above 35.46, respectively. However, digested BC703 using a simulated gastric juice showed poor protective effect against CCl(4)-induced hepatotoxicity in mice and decreased anti-HCV activity as compared to the intact BC703. Although further studies are necessary, BC703 may be a beneficial agent for the management of acute hepatic injury and chronic HCV infection.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Hepacivirus/drug effects , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Platycodon/chemistry , Animals , Hepatitis C/prevention & control , Male , Mice , Mice, Inbred ICR , Phytotherapy , Plant Extracts/chemistry
15.
Food Chem Toxicol ; 50(12): 4254-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22617354

ABSTRACT

Platycodin D (PD) is well known as a potent triterpenoid saponin having various pharmacological activities isolated from the root of Platycodon grandiflorum (Jacq.) A. DC. (Campanulaceae). We aimed to evaluate protective effect of PD on cisplatin (CDDP)-induced nephrotoxicity. Male ICR mice were allocated into five groups as follows: Negative control, CDDP alone and CDDP with PD (0.1, 1 and 5 mg/kg) treated group. PD was given for three consecutive days before CDDP injection. Increased blood urea nitrogen (BUN) and creatinine (CRE) levels in CDDP alone treated mice were decreased to normal range by pretreatment with PD. It also decreased nitric oxide (NO) and lipid peroxidation with increased antioxidant enzymes such as glutathione (GSH), glutathione peroxidase (GPx) and superoxide dismutase (SOD) in PD pretreated mice. In histopathological examination, pretreatment with PD showed ameliorated renal injury such as intraluminal cast formation and epithelial desquamation. Furthermore, over-expression of nuclear factor-kappa B p65 and apoptotic cells were suppressed by PD pretreatment. Taken together, PD pretreatment might be beneficial to CDDP-induced nephrotoxicity.


Subject(s)
Cisplatin/toxicity , Kidney Diseases/drug therapy , Kidney/drug effects , Platycodon/chemistry , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Creatinine/blood , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/pathology , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred ICR , NF-kappa B , Nitric Oxide/blood , Oxidative Stress/drug effects , Plant Roots/chemistry , Superoxide Dismutase/metabolism
16.
Inflammation ; 35(2): 623-32, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21667140

ABSTRACT

Asthma is a complex disease linked to various pathophysiological events, including proteinase activity. In this study, we examined whether a Diospyros blancoi methanolic extract (DBE) exerts protective effects on allergic asthma in a murine asthma model. To investigate the specific role of DBE, we employed a murine model of allergic airway inflammation. BALB/c mice sensitized and challenged with ovalbumin (OVA) were orally administered 20 or 40 mg/kg DBE for 3 days during OVA challenge. DBE induced significant suppression of the number of OVA-induced total inflammatory cells, including eosinophils, macrophages, and lymphocytes, in bronchoalveolar lavage fluid (BALF). Moreover, treatment with DBE led to significant decreases in interleukin (IL)-4, IL-5, and eotaxin levels in BALF and OVA-specific immunoglobulin (Ig)E and IgG1 levels in serum. Histological examination of lung tissue revealed marked attenuation of allergen-induced lung eosinophilic inflammation and mucus-producing goblet cells in the airway. Additionally, DBE suppressed matrix metalloproteinase-9 activity and induced heme oxygenase-1 expression. The present findings collectively suggest that DBE exhibits anti-inflammatory activity in an airway inflammation mouse model, supporting its therapeutic potential for the treatment of allergic bronchial asthma.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Asthma/drug therapy , Diospyros , Plant Extracts/therapeutic use , Animals , Anti-Asthmatic Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Eosinophils/drug effects , Female , Heme Oxygenase-1/metabolism , Immunoglobulin E/blood , Immunoglobulin G/blood , Interleukin-4/metabolism , Interleukin-5/metabolism , Lung/drug effects , Lung/immunology , Lung/pathology , Lymphocytes/drug effects , Macrophages/drug effects , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Phytotherapy , Plant Extracts/pharmacology
17.
Lab Anim Res ; 27(1): 53-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21826161

ABSTRACT

A chordoma is an uncommon tumor that originates from the remnants of the notochord and most commonly involves the cranial and caudal regions of the axial skeleton. Chordoma has been described in laboratory animals such as dogs, rats, minks, and ferrets. This report describes a case of a chordoma in the tail of a ferret. Grossly, a grayish-white, expansile, subcutaneous soft-tissue mass was observed in the tail. Histopathologically, the mass was a loosely placed, nodular, unencapsulated neoplasm within the dermis. In the mass, tumor lobules were intermingled with fibrous tissues. Fibrous tissues contained abundant extracellular basophilic material that was consistent with mucin. The tumor was composed of a close pack of adipocyte-like vacuolated cells (physaliferous cells). The cells were centrally or eccentrically located round nuclei and eosinophilic cytoplasm with large vacuoles. Immunohistologically, neoplastic cells were positive for vimentin and S-100 protein. Based on histopathologic findings and special staining characteristics, this case was diagnosed as chordoma.

18.
Int Immunopharmacol ; 11(6): 698-705, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21295171

ABSTRACT

Chronic airway inflammation is a hallmark of asthma, which is an immune-based disease. We evaluated the ability of saucerneol D, a tetrahydrofuran-type sesquilignan isolated from Saururus chinensis, to regulate airway inflammation in an ovalbumin (OVA)-induced airway inflammation model. Furthermore, we determined whether heme oxygenase (HO)-1 was required for the protective activity of saucerneol D. The airways of OVA-sensitized mice exposed to an OVA challenge developed eosinophilia and mucus hypersecretion and exhibited increased cytokine levels. Mice were administered saucerneol D orally at doses of 20 and 40mg/kg once daily on days 26-30. Saucerneol D administered orally significantly inhibited the number of OVA-induced inflammatory cells and the production of immunoglobulin E as well as Th2-type cytokines. Histopathology studies revealed a marked decrease in lung inflammation and goblet cell hyperplasia after saucerneol D treatment. In addition, saucerneol D induced HO-1 and led to a marked decrease in OVA-induced reactive oxygen species and malondialdehyde and an increase in superoxide dismutase and glutathione in lung tissues. These antioxidant effects were correlated with HO-1 induction. In our experiments, saucerneol D treatment reduced airway inflammation and suppressed oxidative stress in an OVA-induced asthma model.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Antioxidants/administration & dosage , Asthma/drug therapy , Lignans/administration & dosage , Pneumonia/drug therapy , Animals , Anti-Asthmatic Agents/chemistry , Antioxidants/chemistry , Asthma/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Humans , Immunoglobulin E/biosynthesis , Immunoglobulin E/blood , Immunoglobulin E/genetics , Lignans/chemistry , Lignans/pharmacology , Mice , Mice, Inbred BALB C , Pneumonia/chemically induced , Pneumonia/immunology , Reactive Oxygen Species/metabolism , Saururaceae/immunology , Th1-Th2 Balance/drug effects
19.
J Ginseng Res ; 35(2): 243-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-23717067

ABSTRACT

Korean red ginseng (KRG), the steamed root of Panax ginseng Meyer, has a variety of biological properties, including anti-inflammatory, antioxidant and anticancer effects. Aflatoxin B1 (AFB1) produced by the Aspergillus spp. causes acute hepatotoxicity by lipid peroxidation and oxidative DNA damage, and induces liver carcinoma in humans and laboratory animals. This study was performed to examine the protective effects of KRG against hepatotoxicity induced by AFB1 using liver-specific serum marker analysis, histopathology, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling. In addition, to elucidate the possible mechanism of hepatoprotective effects, superoxide dismutase, catalase, glutathione peroxidase, and malondialdehyde were analyzed. Rats were treated with 250 mg/kg of KRG (KRG group) or saline (AFB1 group) for 4 weeks and then received 150 µg/kg of AFB1 intraperitoneally for 3 days. Rats were sacrificed at 12 h, 24 h, 48 h, 72 h, or 1 wk after AFB1 treatment. In the KRG pre-treatment group, serum alanine aminotransferase, aspartate aminotransferase, and malondialdehyde levels were low, but superoxide dismutase, catalase, and glutathione peroxidase activities were high as compared to the AFB1 alone group. Histopathologically, AFB1 treatment induced necrosis and apoptosis in hepatocytes, and led to inflammatory cells infiltration in the liver. KRG pre-treatment ameliorated these changes. These results indicate that KRG may have protective effects against hepatotoxicity induced by AFB1 that involve the antioxidant properties of KRG.

20.
J Ginseng Res ; 35(3): 294-300, 2011 Sep.
Article in English | MEDLINE | ID: mdl-23717072

ABSTRACT

Zearalenone (ZEA) is a phenolic resorcylic acid lactone compound produced by several species of Fusarium. ZEA has toxic effects in the testes of domestic and laboratory animals. Korean red ginseng (KRG), the steamed root of Panax ginseng Meyer, has multiple pharmacological effects such as vasorelaxation, anti-thrombosis, anti-hypertension, etc. In this study, we investigated the effects of KRG extract on testicular toxicity induced by ZEA. Rats were treated with 300 mg/kg oral doses of KRG for 4 weeks every other day. The rats were then treated with a single dose of 5 mg/kg ZEA delivered intraperitoneally, whereas control rats received only doses of the vehicle. As a result, germ cell apoptosis induced by ZEA was decreased by KRG pre-treatment. In addition, Fas and Fas-L expression was reduced in rats that received KRG pre-treatment compared to ones treated with ZEA alone. In conclusion, impaired spermatogenesis resulting from ZEA treatment was prevented by KRG through Fas-Fas L modulating.

SELECTION OF CITATIONS
SEARCH DETAIL
...