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










Publication year range
1.
Exp Ther Med ; 13(5): 2043-2049, 2017 May.
Article in English | MEDLINE | ID: mdl-28565806

ABSTRACT

LD02GIFRO is a novel prokinetic agent formulated with Poncirus fructus and Zanthoxylum sp. fruits. The aim of the present study was to evaluate the effect of LD02GIFRO on delayed gastrointestinal transit (GIT) and colorectal hypersensitivity. To investigate the effect of LD02GIFRO, a rat model of delayed GIT was induced via three mechanisms; postoperative ileus (POI), morphine, and POI plus morphine. Visceromotor responses (VMR) to colorectal distension (CRD) were also evaluated. POI was induced by laparotomy surgery and manipulation of the small intestine under anesthesia, and GIT was calculated by measuring the length that Evans Blue travelled through the gastrointestinal tract in a given time. Oral administration of 260 mg/kg LD02GIFRO caused Evans Blue to migrate significantly further in the delayed GIT models induced by POI, morphine and POI plus morphine compared with the control (P<0.05). This effect was inhibited by atropine, a muscarinic receptor antagonist, and completely abolished by GR125487, a 5-HT4-receptor antagonist. Furthermore, intraperitoneal administration of 600 and 900 mg/kg LD02GIFRO significantly reduced VMR to CRD in acute and chronic colorectal hypersensitive rat models, induced by acetic acid and trinitrobenzenesulfonic acid, to almost normal levels (P<0.01). In the present study, LD02GIFRO successfully ameliorated delayed GIT models and colorectal hypersensitivity models, suggesting that LD02GIFRO may be an effective therapeutic treatment for patients with functional gastrointestinal disorders and abnormalities in GIT.

2.
Mol Med Rep ; 14(5): 4521-4528, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27748812

ABSTRACT

Oxidative stress in liver injury is a major pathogenetic factor in the progression of liver damage. Centella asiatica (L.) Urban, known in the United States as Gotu kola, is widely used as a traditional herbal medicine in Chinese or Indian Pennywort. The efficacy of Centella asiatica is comprehensive and is used as an anti­inflammatory agent, for memory improvement, for its antitumor activity and for treatment of gastric ulcers. The present study investigated the protective effects of Centella asiatica on dimethylnitrosamine (DMN)­induced liver injury in rats. The rats in the treatment groups were treated with Centella asiatica at either 100 or 200 mg/kg in distilled water (D.W) or with silymarin (200 mg/kg in D.W) by oral administration for 5 days daily following intraperitoneal injections of 30 mg/kg DMN. Centella asiatica significantly decreased the relative liver weights in the DMN­induced liver injury group, compared with the control. The assessment of liver histology showed that Centella asiatica significantly alleviated mass periportal ± bridging necrosis, intralobular degeneration and focal necrosis, with fibrosis of liver tissues. Additionally, Centella asiatica significantly decreased the level of malondialdehyde, significantly increased the levels of antioxidant enzymes, including superoxide dismutase, glutathione peroxidase and catalase, and may have provided protection against the deleterious effects of reactive oxygen species. In addition, Centella asiatica significantly decreased inflammatory mediators, including interleukin (IL)­1ß, IL­2, IL­6, IL­10, IL­12, tumor necrosis factor­α, interferon­Î³ and granulocyte/macrophage colony­stimulating factor. These results suggested that Centella asiatica had hepatoprotective effects through increasing the levels of antioxidant enzymes and reducing the levels of inflammatory mediators in rats with DMN­induced liver injury. Therefore, Centella asiatica may be useful in preventing liver damage.


Subject(s)
Centella/chemistry , Chemical and Drug Induced Liver Injury/drug therapy , Inflammation/drug therapy , Triterpenes/administration & dosage , Animals , Antioxidants/metabolism , Catalase/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dimethylnitrosamine/toxicity , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Interleukin-10/metabolism , Liver/drug effects , Liver/pathology , Malondialdehyde/metabolism , Medicine, Chinese Traditional , Oxidative Stress/drug effects , Plant Extracts , Plant Leaves/chemistry , Rats , Superoxide Dismutase/metabolism , Triterpenes/chemistry
3.
Prev Nutr Food Sci ; 21(3): 197-201, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27752495

ABSTRACT

Helicobacter pylori infection is associated with an increased risk of developing upper gastrointestinal tract diseases. However, treatment failure is a major cause of concern mainly due to possible recurrence of infection, the side effects, and resistance to antibiotics. The aim of this study was to investigate the activities of Centella asiatica leaf extract (CAE) against H. pylori both in vitro and in vivo. The minimum inhibitory concentrations (MICs) against 55 clinically isolated strains of H. pylori were tested using an agar dilution method. The MICs of CAE ranged from 0.125 mg/mL to 8 mg/mL, effectiveness in inhibiting H. pylori growth was 2 mg/mL. The anti-H. pylori effects of CAE in vivo were also examined in H. pylori-infected C57BL/6 mice. CAE was orally administrated once daily for 3 weeks at doses of 50 mg/kg and 250 mg/kg. CAE at the 50 mg/kg dose significantly reduced H. pylori colonization in mice gastric mucosa. Our study provides novel insights into the therapeutic effects of CAE against H. pylori infection, and it suggests that CAE may be useful as an alternative therapy.

4.
J Med Food ; 19(1): 38-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26469560

ABSTRACT

The present study evaluated the protective effect of Centella asiatica (gotu kola) leaf extract (CAE) against indomethacin (IND)-induced gastric mucosal injury in rats. Gastric mucosal injury was induced by the oral administration of IND to the rats after a 24 h fast. CAE (50 or 250 mg/kg) or lansoprazole (a reference drug) was orally administrated 30 min before the IND administration, and 5 h later, the stomachs were removed to quantify the lesions. Orally administered CAE significantly reduced IND-induced gastric injury. The histopathological observations (hematoxylin-eosin and Periodic acid-Schiff staining) confirmed the protection against gastric mucosal injury. Also, CAE decreased the malondialdehyde content compared to the control group. Moreover, pretreatment with CAE resulted in a significant reduction in the elevated expression of tumor necrosis factor, Cyclooxygenase (COX)-2, and inducible nitric oxide synthase. These results suggested that CAE possesses gastroprotective effects against IND-induced gastric mucosal injury, which could be attributed to its ability to inhibit lipid peroxidation and stimulate gastric mucus secretion in the rat gastric mucosa.


Subject(s)
Centella/chemistry , Gastric Mucosa/drug effects , Indomethacin/administration & dosage , Plant Extracts/administration & dosage , Stomach Ulcer/drug therapy , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gastric Mucosa/injuries , Gastric Mucosa/metabolism , Humans , Male , Malondialdehyde/metabolism , Plant Leaves/chemistry , Rats , Rats, Sprague-Dawley , Stomach Ulcer/chemically induced , Stomach Ulcer/genetics , Stomach Ulcer/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Cancer Lett ; 332(1): 74-82, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23348694

ABSTRACT

Among many cancer therapeutic targets, c-Met receptor tyrosine kinase has recently given particular attention. This kinase and its ligand, hepatocyte growth factor (HGF), play a central role in cell proliferation and the survival of several human cancers. Thus, we developed KRC-408 as a novel c-Met inhibitor and investigated its anti-cancer effects on human gastric cancer. KRC-408 inhibited the phosphorylation of c-Met and its constitutive downstream effectors such as phosphatidylinositol 3-kinase (PI3K), Akt, Mek, and Erk. This compound was found to exert anti-cancer effects stronger than those of 5-fluorouracil (5-FU) on gastric cancer cells, especially cell lines that overexpressed c-Met. Interestingly, cytotoxicity of KRC-408 was lower than that of 5-FU in normal gastric cells. Apoptosis induced by KRC-408 was accompanied by increased levels of cleaved caspase-3 and PARP as well as DNA condensation and fragmentation. Flow cytometry analysis showed an accumulation of gastric cancer cells in the G2/M phase with concomitant loss of cells in the S phase following treatment with this drug. In the angiogenesis studies, KRC-408 inhibited tube formation and migration of human umbilical vein endothelial cells (HUVECs), and suppressed microvessel sprouting from rat aortic rings ex vivo along with blood vessel formation in a Matrigel plug assay in mice. Results of an in vivo mouse xenograft experiment showed that the administration of KRC-408 significantly delayed tumor growth in a dose-dependent manner, and suppressed Akt and Erk phosphorylation as well CD34 expression in tumor tissues. These findings indicate that KCR-408 may exert anti-tumor effects by directly affecting tumor cell growth or survival via the c-Met receptor tyrosine kinase pathway. We therefore suggest that KRC-408 is a novel therapeutic candidate effective against gastric cancers that overexpress c-Met.


Subject(s)
Aminopyridines/pharmacology , Angiogenesis Inhibitors/pharmacology , Benzoxazoles/pharmacology , Cell Proliferation/drug effects , Neovascularization, Physiologic/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Aminopyridines/chemistry , Angiogenesis Inhibitors/chemistry , Animals , Antigens, CD34/metabolism , Apoptosis/drug effects , Benzoxazoles/chemistry , Caspase 3/metabolism , Cell Line, Tumor , DNA Fragmentation , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Molecular Structure , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stomach Neoplasms/blood supply , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Time Factors , Tissue Culture Techniques , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Cancer Lett ; 328(1): 176-87, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22902995

ABSTRACT

Dysregulation of the phosphoinositide 3-kinase (PI3K)/AKT/mTOR signaling pathway frequently instigates tumorigenesis leading to hepatocellular carcinoma (HCC). We synthesized N-(5-(3-(3-methyl-1,2,4-oxadiazol-3-yl)imidazo[1,2-a]pyridin-6-yl)pyridin-3-yl)benzenesulfonamide (HS-104), a novel PI3K inhibitor, and investigated its in vitro anticancer effect and in vivo capacity in an animal xenograft model. The inhibition of cell growth by HS-104 revealed that it was effective against HCC cell lines. Also, the activation of the AKT/mTOR signal cascade was inhibited by HS-104 treatment in a dose dependent manner. Flow cytometry analysis showed an accumulation of HCC cells in the G2/M phase with concomitant loss of cells in the S phase. The apoptotic effect of HS-104 was accompanied by increased evidence of cleaved caspase-3 and PARP, as well as DNA fragmentation. In angiogenesis studies, HS-104 inhibited the tube formation of vascular endothelial growth factor (VEGF)-induced human umbilical vein endothelial cells (HUVECs), and suppressed microvessel sprouting from a rat aortic ring, ex vivo, and blood vessel formation in the Matrigel plug assay in mice. HS-104 inhibited the expression of the downstream proteins of PI3K including p-AKT, p-mTOR and p-p70S6K in VEGF-induced HUVECs. In the xenograft animal model, HS-104 significantly delayed tumor growth in a dose dependent manner and suppressed the expression of PCNA, CD34 and cleaved caspase-3 in tumor tissue. These studies show that HS-104 inhibited the PI3K/AKT/mTOR signaling pathway resulting in cell growth/angiogenesis inhibition and apoptosis induction. Therefore, HS-104 is considered as a novel drug candidate for the treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Imidazoles/pharmacology , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Pyridines/pharmacology , Animals , Carcinoma, Hepatocellular/blood supply , Cell Line, Tumor , Humans , Liver Neoplasms/blood supply , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
7.
Food Chem ; 136(1): 26-33, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23017388

ABSTRACT

Pulsatilla koreana has been used as a traditional medicine for the treatment of several diseases. The purpose of this study was to determine if SB365, Pulsatilla saponin D isolated from the root of P. koreana inhibits the progression of colon cancer. We found that SB365 strongly suppressed the growth and proliferation of colon cancer cells and induced their apoptosis. Also, SB365 showed anti-angiogenic activity by decreasing the expression of HIF-1α and VEGF. These results were confirmed by an in vivo study showing that SB365 significantly inhibited tumor growth by the induction of apoptosis and inhibition of angiogenesis with stronger anticancer activity than 5-FU. When further examined for its anticancer mechanism, SB365 effectively suppressed the AKT/mTOR pathway both in vitro and in vivo. Taken together, our study demonstrated that SB365 inhibits the AKT/mTOR pathway, leading to the suppression of tumor growth and angiogenesis together with induction of apoptosis. Therefore, SB365 is a good candidate as a natural product for use in the treatment of colon cancer.


Subject(s)
Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pulsatilla/chemistry , Saponins/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/physiopathology , Down-Regulation/drug effects , Humans , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics
8.
Int J Oncol ; 41(5): 1715-22, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22941304

ABSTRACT

The anticancer effect of a new pyrazole derivative, KI-10F (2-(4-(2-(4-(dimethylamino) phenyl)pyridin-4-yl)-5-(3-methoxy-5-methylphenyl)-1H-pyrazol­1-yl) acetonitrile)•3.5HCl) was evaluated in human colon cancer cells. KI-10F strongly suppressed the growth of human colon cancer cells and induced apoptosis by increasing the proportion of sub-G1 presenting apoptotic cells as well as causing cell cycle arrest at the G2/M phase. Apoptosis by KI-10F was confirmed by observation of an increase in the expression of cleaved caspase-3, caspase-8, caspase-9 and Bax, and the decrease of Bcl-2. Decreased expression of HIF-1α and VEGF, and the inhibition of HUVEC tube formation and migration showed that KI-10F effectively inhibited the angiogenesis process. Furthermore, in vivo study in a mouse xenograft model showed that KI-10F produced a stronger antitumor activity than 5-FU, a conventional anticancer drug prescribed for the treatment of colon cancer. The effects of KI-10F on tumor proliferation (PCNA), angiogenesis (CD34) and apoptosis (cleaved caspase-3) were evaluated by immunohistochemistry using isolated tumor tissue samples. Taken together, our results demonstrated that KI-10F induces apoptosis and inhibits cell growth and angiogenesis both in vitro and in vivo. We suggest that KI-10F is an effective chemotherapeutic candidate for use against colon cancer.


Subject(s)
Acetonitriles/pharmacology , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/metabolism , Neovascularization, Pathologic , Pyrazoles/pharmacology , Acetonitriles/administration & dosage , Acetonitriles/chemistry , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , HT29 Cells , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Cancer Sci ; 103(11): 1929-37, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22909393

ABSTRACT

Identification of small molecules that safely inhibit cancer progression is critical for cancer therapeutics. Saponins exhibit cytostatic and cytotoxic activity against various cancer cells, but the mechanism is not well understood. Here, we investigated whether saponin D (designated SB365), an active component isolated from Pulsatilla koreana, could inhibit the progression of hepatocellular carcinoma (HCC) and considered its mechanism. SB365 strongly suppressed the growth of HCC cells in a dose-dependent manner and induced apoptosis by increasing the proportion of sub G1 apoptotic cells from 8% to 21% through induction of expression of Bax and cleaved caspase-3. In addition, SB365 exhibited potent anti-angiogenic activity and decreased the expression of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor, a key molecule for angiogenesis. Furthermore, SB365 suppressed the tube formation and migration of HUVEC, as well as in vivo neovascularization in a mouse Matrigel plug assay. In vivo study showed that SB365 significantly inhibited tumor growth in an HCC xenograft model, inducing apoptosis by increasing the expression of the cleaved caspase-3 and DNA fragmentation. The expressions of vascular endothelial growth factor and CD34 in the tumor tissue were decreased by SB365 treatment. In examining its mechanism, SB365 was found to effectively suppress the phosphorylation of PI3K downstream factors, such as Akt, mTOR and p70S6K both in vitro and in vivo. Our study demonstrates that SB365 not only induces apoptosis but also inhibits cell growth and angiogenesis through modulation of the PI3K/Akt/mTOR pathway in human HCC. We suggest that SB365 may be a new chemotherapeutic candidate against HCC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Saponins/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
10.
Arch Pharm Res ; 35(4): 747-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22553069

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies, yet there have been no significant advances in effective therapeutics. In this study, HS-111 was synthesized as a novel thiazolamine derivative, N-(5-(2-chlorobenzyl) thiazole-2-yl) benzofuran-2-carboxamide, and its anticancer effect and mechanism were examined in human HCC cells. HS-111 strongly suppressed the growth of HCC cells in a dose-dependent manner. Also, apoptosis by HS-111 was identified by DAPI and TUNEL staining, and the increases of the cleaved caspase-3 were observed. In addition, HS-111 decreased protein expression of hypoxia-inducible factor (HIF-1α) and secretion of vascular endothelial growth factor (VEGF), and inhibited tube formation and the migration of human umbilical vein endothelial cells (HUVECs). These results showed that HS-111 not only inhibited cell growth and angiogenesis, but also induced apoptosis of human HCC cells. We suggest that HS-111 may be a potential candidate for chemotherapy against HCC.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Benzofurans/pharmacology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Neovascularization, Pathologic/prevention & control , Thiazoles/chemistry , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Benzofurans/chemistry , Benzofurans/therapeutic use , Blotting, Western , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells , Humans , In Situ Nick-End Labeling , Liver Neoplasms/blood supply , Liver Neoplasms/drug therapy , Molecular Structure , Thiazoles/pharmacology , Thiazoles/therapeutic use , Wound Healing/drug effects
11.
Int J Oncol ; 40(2): 452-60, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21935571

ABSTRACT

Chemoprevention through the use of food and plants has emerged as a novel approach to control various malignancies including cancer. Pulsatilla koreana extract (PKE) has been used to treat malaria and dysentery. The functions and effect of PKE in cancer treatment have been reported but with less information. In this study, we investigated the effect of PKE on the progression of hepatocellular carcinoma (HCC) cells and its mechanism. PKE strongly suppressed the growth of HCC cells in a dose-dependent manner. Apoptosis by PKE was observed by DAPI and TUNEL staining and accompanied with increases of cleaved PARP and caspase-3 in Huh-7 cells. Also, PKE decreased the expression of hypoxia-inducible factor (HIF-1α) and vascular endothelial growth factor (VEGF), and inhibited tube formation and migration of human umbilical vein endothelial cells (HUVECs). In addition, PKE potently suppressed in vivo neovascularization in a mouse Matrigel plug assay. Furthermore, in vivo study showed that PKE significantly inhibited tumor growth in a mouse xenograft model, and induced apoptosis by increasing the cleaved PARP and caspase-3. The expressions of Ki-67, VEGF, and CD31 in the tumor tissue were decreased by the treatment of PKE. Taken together, our study demonstrates that PKE not only induced apoptosis but also inhibited cell growth and angiogenesis of human HCC. We suggest that PKE is an effective chemotherapeutic candidate for cancer therapy against HCC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Plant Extracts/pharmacology , Plant Roots , Pulsatilla , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/urine , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Plant Extracts/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Lett ; 316(2): 187-95, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22182943

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) pathway plays a central role in cell proliferation and survival of human cancers. As PI3K is active in many cancer patients, resulting in cancer development and progression, we developed an azaindole derivative, HS-116 as a novel PI3K inhibitor. This study aimed to clarify the anticancer effect of HS-116 in human hepatocellular carcinoma (HCC). To identify the effect of HS-116 on HCC cells, a PI3K assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, flow cytometry, and Western blotting were conducted. IC(50) of HS-116 for PI3Kα was 31nM, and it effectively suppressed the phosphorylation of PI3K downstream factors such as AKT, mTOR, p70S6K, and 4EBP1. Also, HS-116 induced apoptosis by increasing the proportion of sub-G1 apoptotic cells from 1.8% to 35% and increasing the expressions of Bax, cleaved-caspase-3, and cleaved-PARP as well as decreasing the expression of Bcl-2. In addition, chromatin condensation and apoptotic bodies were detected in HS-116-treated HCC cells. Furthermore, HS-116 decreased protein expression of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF), and inhibited the tube formation and migration of human umbilical vein endothelial cells (HUVECs). In vivo, the ability of mice to vascularize subcutaneously implanted Matrigel plugs was diminished when the mice were treated with HS-116. These results show that HS-116 inhibits the PI3K/AKT/mTOR pathway via apoptosis and anti-angiogenesis in HCC cells. We suggest that HS-116 may be an effective novel therapeutic candidate against HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/pharmacology , Pyrroles/pharmacology , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Hep G2 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
13.
Biosci Biotechnol Biochem ; 75(5): 833-40, 2011.
Article in English | MEDLINE | ID: mdl-21597183

ABSTRACT

Fucoidan, a sulfated polysaccharide extracted from various types of brown seaweed, possesses a wide range of pharmacological properties. We investigated the protective effect of fucoidan on dimethylnitrosamine-induced liver fibrogenesis in rats and its mechanism. Liver fibrosis was induced by injecting DMN (10 mg/kg, 3 times per week, I.P.) for 4 weeks, and fucoidan was simultaneously administered (100 mg/kg, 3 times per week, P.O.). The anti-oxidative and anti-inflammatory effects of fucoidan were observed by relative mediators. Fucoidan improved liver fibrosis by inhibiting the expression of transforming growth factor beta 1 (TGF-ß(1))/Smad3 and the tissue inhibitor of metalloproteinase 1 (TIMP-1), and increasing the expression of metalloproteinase-9 (MMP-9). Fucoidan also significantly decreased the accumulation of the extracellular matrix (ECM) and collagen. These results suggest that fucoidan had an anti-fibrotic effect, which was exerted by inhibiting the TGF-ß/Smad pathway, as well as anti-oxidative and anti-inflammatory effects.


Subject(s)
Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Oxidative Stress/drug effects , Polysaccharides/pharmacology , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antioxidants/metabolism , Body Weight/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Glutathione Peroxidase/blood , Glutathione Peroxidase/metabolism , Inflammation Mediators/metabolism , Liver Cirrhosis/blood , Liver Cirrhosis/metabolism , Male , Malondialdehyde/blood , Malondialdehyde/metabolism , Organ Size/drug effects , Polysaccharides/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/blood , Superoxide Dismutase/metabolism
14.
Cancer Lett ; 306(2): 190-6, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21463918

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies, yet there have been no significant advances in effective therapeutics. In this study, HS-113 was synthesized as a novel compound, N-(5-(2-bromobenzyl) thiazole-2-yl) benzofuran-2-carboxamide and its cytotoxic activity and anti-cancer effect were examined in human HCC cells. HS-113 strongly suppressed growth of HCC cells in a dose-dependent manner, induced apoptosis by increasing the proportion of sub-G1 apoptotic cells, and caused cell cycle arrest at G0/G1 phase. Also, HS-113 increased the expression of p27 and decreased that of cyclin D1 associated with cell cycle arrest. Apoptosis by HS-113 was confirmed by DAPI and TUNEL staining, and the increases of the cleaved PARP and caspase-3 were observed. Furthermore, HS-113 decreased protein expression of HIF-1α and secretion of VEGF, and inhibited the tube formation of HUVECs. These results showed that HS-113 not only inhibited cell growth and angiogenesis, but also induced apoptosis of human HCC cells. We suggest that HS-113 may be a potential candidate for cancer therapy against HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzofurans/pharmacology , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Neovascularization, Pathologic/prevention & control , Thiazoles/pharmacology , Benzofurans/chemical synthesis , Blotting, Western , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Thiazoles/chemical synthesis , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism
15.
PLoS One ; 6(2): e16641, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21364936

ABSTRACT

BACKGROUND: Simvastatin, which is used to control elevated cholesterol levels, is one of the most widely prescribed drugs. However, a daily excessive dose can induce drug-toxicity, especially in muscle and liver. Current markers for toxicity reflect mostly the late stages of tissue damage; thus, more efficient methods of toxicity evaluation are desired. METHODOLOGY/PRINCIPAL FINDINGS: As a new way to evaluate toxicity, we performed NMR-based metabonomics analysis of urine samples. Compared to conventional markers, such as AST, ALT, and CK, the urine metabolic profile provided clearer distinction between the pre- and post-treatment groups treated with toxic levels of simvastatin. Through multivariate statistical analysis, we identified marker metabolites associated with the toxicity. Importantly, we observed that the treatment group could be further categorized into two subgroups based on the NMR profiles: weak toxicity (WT) and high toxicity (HT). The distinction between these two groups was confirmed by the enzyme values and histopathological exams. Time-dependent studies showed that the toxicity at 10 days could be reliably predicted from the metabolic profiles at 6 days. CONCLUSIONS/SIGNIFICANCE: This metabonomics approach may provide a non-invasive and effective way to evaluate the simvastatin-induced toxicity in a manner that can complement current measures. The approach is expected to find broader application in other drug-induced toxicity assessments.


Subject(s)
Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions/diagnosis , Metabolomics/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Simvastatin/adverse effects , Animals , Biomarkers, Pharmacological/blood , Biomarkers, Pharmacological/metabolism , Biomarkers, Pharmacological/urine , Blood Chemical Analysis/methods , Drug Evaluation, Preclinical/instrumentation , Female , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Metabolome/drug effects , Metabolomics/instrumentation , Nuclear Magnetic Resonance, Biomolecular/instrumentation , Rats , Rats, Wistar , Simvastatin/pharmacology , Treatment Outcome , Urinalysis/methods
16.
Kidney Int ; 79(5): 529-37, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20980974

ABSTRACT

Cisplatin has been one of the most widely used anticancer agents, but its nephrotoxicity remains a dose-limiting complication. Here, we evaluated the idiopathic nature and the predose prediction of cisplatin-induced nephrotoxicity using a nuclear magnetic resonance (NMR)-based pharmacometabonomic approach. Cisplatin produced serious toxic responses in some animals (toxic group), but had little effect in others (nontoxic group), as judged by hematological and histological results. The individual metabolic profiles, assessed by urine NMR spectra, showed large differences between the post-administration profiles of the two groups, indicating the relevance of the NMR approach. Importantly, multivariate analysis of the NMR data showed that the toxic and nontoxic groups can be differentiated based on the pretreatment metabolite profiles. Leave-one-out analysis, performed to evaluate the practical performance of our approach, gave a 66% accuracy rate in predicting toxic responses based on the pretreatment metabolite profiles. Hence, we provide a working model that can explain the idiopathic toxicity mechanism based on marker metabolites found by NMR analysis consistent with tissue NADH measurements. Thus, a pharmacometabonomic approach using pretreatment metabolite profiles may help expedite personalized chemotherapy of anticancer drugs.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney/drug effects , Animals , Kidney/metabolism , Magnetic Resonance Spectroscopy , Male , Metabolomics , Multivariate Analysis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
17.
Gastroenterology ; 140(3): 998-1008, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21130088

ABSTRACT

BACKGROUND & AIMS: Acute pancreatitis (AP) has a high mortality rate; repetitive AP induces chronic AP and pancreatic adenocarcinoma. Mesenchymal stem cells (MSCs) have immunoregulatory effects and reduce inflammation. We developed a protocol to isolate human bone marrow-derived clonal MSCs (hcMSCs) from bone marrow aspirate and investigated the effects of these cells in rat models of mild and severe AP. METHODS: Mild AP was induced in Sprague-Dawley rats by 3 intraperitoneal injections of cerulein (100 µg/kg), given at 2-hour intervals; severe AP was induced by intraparenchymal injection of 3% sodium taurocholate solution. hcMSCs were labeled with CM-1,1'-dioctadecyl-3,3,3'-tetramethylindo-carbocyanine perchloride and administered to rats through the tail vein. RESULTS: hcMSCs underwent self-renewal and had multipotent differentiation capacities and immunoregulatory functions. Greater numbers of infused hcMSCs were detected in pancreas of rats with mild and severe AP than of control rats. Infused hcMSCs reduced acinar-cell degeneration, pancreatic edema, and inflammatory cell infiltration in each model of pancreatitis. The hcMSCs reduced expression of inflammation mediators and cytokines in rats with mild and severe AP. hcMSCs suppressed the mixed lymphocyte reaction and increased expression of Foxp3(+) (a marker of regulatory T cells) in cultured rat lymph node cells. Rats with mild or severe AP that were given infusions of hcMSCs had reduced numbers of CD3(+) T cells and increased expression of Foxp3(+) in pancreas tissues. CONCLUSIONS: hcMSCs reduced inflammation and damage to pancreatic tissue in a rat model of AP; they reduced levels of cytokines and induced numbers of Foxp3(+) regulatory T cells. hcMSCs might be developed as a cell therapy for pancreatitis.


Subject(s)
Bone Marrow Transplantation , Mesenchymal Stem Cell Transplantation , Pancreas/surgery , Pancreatitis/surgery , Acute Disease , Animals , Biomarkers/metabolism , CD3 Complex/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Ceruletide , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , In Situ Hybridization, Fluorescence , Inflammation Mediators/metabolism , Pancreas/immunology , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/immunology , Pancreatitis/metabolism , Pancreatitis/pathology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Regeneration , Severity of Illness Index , T-Lymphocytes/immunology , Taurocholic Acid , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...