Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Int J Mol Sci ; 19(2)2018 Feb 12.
Article in English | MEDLINE | ID: mdl-29439518

ABSTRACT

Cinnamic aldehyde (CA), a key flavor compound in cinnamon essential oil, has been identified as an anti-oxidant, anti-angiogenic, and anti-inflammatory material. Recently, the neuroprotective effects of CA have been reported in various neurodegenerative disorders, including Parkinson's disease (PD). In neurons, autophagy is tightly regulated, and consequently, the dysregulation of autophagy may induce neurodegenerative disorders. In the present study, we found that the selective dopaminergic neuronal death in the substantia nigra of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse models was prevented by CA. Stimulation of microtubule-associated protein light chain 3 (LC3) puncta mediated by MPTP treatment was decreased by CA. Moreover, down-regulated p62 in the substantia nigra of MPTP mice was increased by administration of CA. Finally, we showed that blockage of autophagy using autophagy inhibitors protected the 1-methyl-4-phenylpyridinium (MPP⁺)-mediated death of BE(2)-M17 cells. Together these results suggest that CA has a neuroprotective effect in a PD model and that inhibition of autophagy might be a promising therapeutic target for PD.


Subject(s)
Acrolein/analogs & derivatives , MPTP Poisoning/drug therapy , Neuroprotective Agents/therapeutic use , Acrolein/pharmacology , Acrolein/therapeutic use , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism
2.
Int J Mol Sci ; 19(8)2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30081604

ABSTRACT

Hypoxia-inducible factors (HIFs) are key regulators of hypoxic responses, and their stability and transcriptional activity are controlled by several kinases. However, the regulation of HIF by protein phosphatases has not been thoroughly investigated. Here, we found that overexpression of Mg2+/Mn2+-dependent protein phosphatase 1 gamma (PPM1G), one of Ser/Thr protein phosphatases, downregulated protein expression of ectopic HIF-1α under normoxic or acute hypoxic conditions. In addition, the deficiency of PPM1G upregulated protein expression of endogenous HIF-1α under normoxic or acute oxidative stress conditions. PPM1G decreased expression of HIF-1α via the proteasomal pathway. PPM1G-mediated HIF-1α degradation was dependent on prolyl hydroxylase (PHD), but independent of von Hippel-Lindau (VHL). These data suggest that PPM1G is critical for the control of HIF-1α-dependent responses.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Phosphatase 2C/metabolism , Blotting, Western , Cell Hypoxia/genetics , Cell Hypoxia/physiology , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Oxidative Stress/genetics , Oxidative Stress/physiology , Protein Binding , Protein Phosphatase 2C/genetics , Reverse Transcriptase Polymerase Chain Reaction , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
3.
Int J Mol Sci ; 19(9)2018 Sep 19.
Article in English | MEDLINE | ID: mdl-30235818

ABSTRACT

Cancer cells undergo uncontrolled proliferation resulting from aberrant activity of various cell-cycle proteins. Therefore, despite recent advances in intensive chemotherapy, it is difficult to cure cancer completely. Recently, cell-cycle regulators became attractive targets in cancer therapy. Zingerone, a phenolic compound isolated from ginger, is a nontoxic and inexpensive compound with varied pharmacological activities. In this study, the therapeutic effect of zingerone as an anti-mitotic agent in human neuroblastoma cells was investigated. Following treatment of BE(2)-M17 cells with zingerone, we performed a 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide (MTT) assay and colony-formation assay to evaluate cellular proliferation, in addition to immunofluorescence cytochemistry and flow cytometry to examine the mitotic cells. The association of gene expression with tumor stage and survival was analyzed. Furthermore, to examine the anti-cancer effect of zingerone, we applied a BALB/c mouse-tumor model using a BALB/c-derived adenocarcinoma cell line. In human neuroblastoma cells, zingerone inhibited cellular viability and survival. Moreover, the number of mitotic cells, particularly those in prometaphase, increased in zingerone-treated neuroblastoma cells. Regarding specific molecular mechanisms, zingerone decreased cyclin D1 expression and induced the cleavage of caspase-3 and poly (ADP-ribose) polymerase 1 (PARP-1). The decrease in cyclin D1 and increase in histone H3 phosphorylated (p)-Ser10 were confirmed by immunohistochemistry in tumor tissues administered with zingerone. These results suggest that zingerone induces mitotic arrest followed by inhibition of growth of neuroblastoma cells. Collectively, zingerone may be a potential therapeutic drug for human cancers, including neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin D1/genetics , Guaiacol/analogs & derivatives , M Phase Cell Cycle Checkpoints/drug effects , Mitosis/drug effects , Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Caspase 3/metabolism , Cell Line, Tumor , Cyclin D1/metabolism , Guaiacol/pharmacology , Guaiacol/therapeutic use , Humans , Male , Mice , Mice, Inbred BALB C , Poly (ADP-Ribose) Polymerase-1/metabolism
4.
Biophys J ; 111(8): 1724-1737, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27760359

ABSTRACT

There is a significant need for developing compounds that kill Cryptococcus neoformans, the fungal pathogen that causes meningoencephalitis in immunocompromised individuals. Here, we report the mode of action of a designed antifungal peptide, VG16KRKP (VARGWKRKCPLFGKGG) against C. neoformans. It is shown that VG16KRKP kills fungal cells mainly through membrane compromise leading to efflux of ions and cell metabolites. Intracellular localization, inhibition of in vitro transcription, and DNA binding suggest a secondary mode of action for the peptide, hinting at possible intracellular targets. Atomistic structure of the peptide determined by NMR experiments on live C. neoformans cells reveals an amphipathic arrangement stabilized by hydrophobic interactions among A2, W5, and F12, a conventional folding pattern also known to play a major role in peptide-mediated Gram-negative bacterial killing, revealing the importance of this motif. These structural details in the context of live cell provide valuable insights into the design of potent peptides for effective treatment of human and plant fungal infections.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Cryptococcus neoformans/drug effects , Active Transport, Cell Nucleus , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Base Sequence , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cryptococcus neoformans/cytology , DNA/chemistry , DNA/genetics , DNA/metabolism , Models, Molecular , Nucleic Acid Conformation
5.
Phytother Res ; 29(11): 1783-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26282055

ABSTRACT

Parkinson's disease (PD) is characterized by a progressive and selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and striatum. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is used to produce an animal model for PD, and it is converted to 1-methyl-4-phenylpyridine (MPP(+)) in animals. MPP(+) accumulation leads to neuronal cell death. Vesicular monoamine transporter 2 (VMAT2) regulates the accumulation of monoamine neurotransmitters into synaptic vesicles and is involved in neuroprotection against neurotoxin-induced cell death. Recently, zingerone has been reported to reduce oxidative stress and inhibit inflammation. Therefore, we examined the effect of zingerone on neuronal cell death in a PD model. In an MPP(+) and MPTP-mediated PD model, neuronal cell survival was increased by zingerone without modifying neuroinflammation or reactive oxygen species generation. Zingerone also induced ERK activation and VMAT2 expression, leading to the attenuation of MPP(+)-induced neuronal cell death. Our current results suggest that zingerone has a neuroprotective effect in a PD model.


Subject(s)
Cell Death/drug effects , Guaiacol/analogs & derivatives , Vesicular Monoamine Transport Proteins/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Corpus Striatum/drug effects , Disease Models, Animal , Dopaminergic Neurons/drug effects , Drug Interactions , Guaiacol/pharmacology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neurotoxins , Parkinson Disease/drug therapy
6.
Phytother Res ; 28(8): 1246-51, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24535656

ABSTRACT

Salicin has been studied as a potent antiinflammatory agent. Angiogenesis is an essential process for tumor progression, and negative regulation of angiogenesis provides a good strategy for antitumor therapy. However, the potential medicinal value of salicin on antitumorigenic and antiangiogenic effects remain unexplored. In this study, we examined the antitumorigenic and antiangiogenic activity of salicin and its underlying mechanism of action. Salicin suppressed the angiogenic activity of endothelial cells, such as migration, tube formation, and sprouting from an aorta. Moreover, salicin reduced reactive oxygen species production and activation of the extracellular signal-regulated kinase pathway. The expression of vascular endothelial growth factor was also decreased by salicin in endothelial cells. When the salicin was administered to mice, salicin inhibited tumor growth and angiogenesis in a mouse tumor model. Taken together, salicin targets the signaling pathways mediated by reactive oxygen species and extracellular signal-regulated kinase, providing new perspectives into a potent therapeutic agent for hypervascularized tumors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Benzyl Alcohols/pharmacology , Glucosides/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , MAP Kinase Signaling System/drug effects , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Aorta/drug effects , Cell Line, Tumor , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Plant Bark/chemistry , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Salix/chemistry
7.
J Pineal Res ; 54(3): 264-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22924616

ABSTRACT

The sustained expansion of a tumor mass requires new blood vessel formation to provide rapidly proliferating tumor cells with an adequate supply of oxygen and nutrients. Hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor angiogenesis and growth by regulating the transcription of genes in response to hypoxic stress. This study was designed to investigate the effects of melatonin on tumor growth and angiogenesis, as well as the mechanism underlying the antitumor activities of melatonin. In this study, we show that the administration of melatonin inhibits tumor growth and blocks tumor angiogenesis in mice. Moreover, melatonin diminished the expression of the HIF-1α protein within the tumor mass during tumorigenesis. Our findings suggest that melatonin is a promising anti-angiogenic therapeutic agent targeting HIF-1α in cancer. Considering that HIF-1α is overexpressed in a majority of human cancers, melatonin could offer a potent therapeutic agent for cancer.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Melatonin/pharmacology , Animals , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Microvessels/drug effects , Microvessels/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
8.
Phytother Res ; 27(6): 841-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22899320

ABSTRACT

Glycyrrhizic acid (GA) is the bioactive compound of licorice and has been used as a herbal medicine because of its anti-viral, anti-cancer, and anti-inflammatory properties. This study was designed to investigate the effects of GA on tumor growth, angiogenesis, and the mechanisms underlying the anti-angiogenic activities of GA. We observed that GA inhibited tumor growth and angiogenesis in mice. GA decreased angiogenic activities, such as the migration, invasion, and tube formation of endothelial cells. We also demonstrated that GA reduced the production of reactive oxygen species and activation of ERK in endothelial cells. Our findings suggest that GA is a promising anti-angiogenic therapeutic agent that targets the ERK pathway. Considering that angiogenesis is highly stimulated in the majority of cancers, GA could offer a potent therapeutic agent for cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Endothelial Cells/drug effects , Glycyrrhizic Acid/pharmacology , Animals , Cell Line, Tumor , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
9.
Phytother Res ; 27(3): 317-23, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22585412

ABSTRACT

p-Coumaric acid, a hydroxy derivative of cinnamic acid, has been known to possess antioxidant and anticancer activities. Despite its potential contribution to chemopreventive effects, the mechanism by which p-coumaric acid exerts its antiangiogenic actions remains elusive. In this study, we revealed that p-coumaric acid inhibited the sprouting of endothelial cells in rat aortic rings and inhibited the tube formation and migration of endothelial cells. We observed that p-coumaric acid could downregulate mRNA expression levels of the key angiogenic factors vascular endothelial growth factor and basic fibroblast growth factor. Also, we demonstrated that p-coumaric acid inhibited both the AKT and ERK signaling pathways, which are known to be crucial for angiogenesis. Using a mouse model, we also showed that p-coumaric acid effectively suppressed tumor growth in vivo by lowering hemoglobin contents. Collectively, these findings indicate that p-coumaric acid possesses potent anticancer properties due to the inhibition of angiogenesis in vivo.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Coumaric Acids/pharmacology , Endothelial Cells/drug effects , Neovascularization, Pathologic/drug therapy , Adenocarcinoma/blood supply , Animals , Antineoplastic Agents/pharmacology , Aorta/cytology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Fibroblast Growth Factor 2/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Propionates , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
10.
Environ Pollut ; 329: 121715, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37120000

ABSTRACT

Fine particulate matter (PM2.5) is associated with public health problems worldwide. Especially, PM2.5 induces epigenetic and microenvironmental changes in lung cancer. Angiogenesis is important for the development and growth of cancer and is mediated by angiogenic factors, including vascular endothelial growth factor. However, the effects of mild PM2.5 exposure on angiogenesis in lung cancer remain unclear. In this study, we examined angiogenic effects using relatively lower concentrations of PM2.5 than in other studies and found that PM2.5 increased angiogenic activities in both endothelial cells and non-small cell lung carcinoma cells. PM2.5 also promoted the growth and angiogenesis of lung cancer via the induction of hypoxia-inducible factor-1α (HIF-1α) in a xenograft mouse tumor model. Angiogenic factors, including vascular endothelial growth factor (VEGF), were highly expressed in lung cancer patients in countries with high PM2.5 levels in the atmosphere, and high expression of VEGF in lung cancer patients lowered the survival rate. Collectively, these results provide new insight into the mechanisms by which mild exposure to PM2.5 is involved in HIF-1α-mediated angiogenesis in lung cancer patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Vascular Endothelial Growth Factor A/metabolism , Particulate Matter/toxicity , Endothelial Cells/metabolism , Cell Line, Tumor
11.
J Cell Biochem ; 113(12): 3730-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22786746

ABSTRACT

Cells under hypoxic stress either activate an adaptive response or undergo cell death. Although some mechanisms have been reported, the exact mechanism behind hypoxic cell death remains unclear. Recently, increased expression of fatty acid synthase (FASN) has been observed in various human cancers. In highly proliferating cells, tumor-associated FASN is considered necessary for both membrane lipids production and post-translational protein modification, but the exact mechanisms are not fully understood. Further, FASN overexpression is associated with aggressive and malignant cancer diseases and FASN inhibition induces apoptosis in cancer cells. For this reason, FASN is emerging as a key target for the potential diagnosis and treatment of various cancers. Here, we observed decreased FASN expression under hypoxic cell death conditions in HepG2 cells. Thus, we examined the effect of decreased FASN expression on hypoxia-induced cell death in HepG2 cells and also investigated the mechanism responsible for reduction of FASN expression under hypoxic cell death conditions. As a result, reduction of FASN expression resulted in hypoxic cell death via malonyl-CoA accumulation. In addition, SREBP-1 restored FASN reduction and hypoxia-induced apoptosis. Taken together, we suggest that hypoxic cell death is promoted by the reduced expression of FASN through SREBP-1 down-regulation.


Subject(s)
Down-Regulation , Fatty Acid Synthase, Type I/metabolism , Gene Expression Regulation, Neoplastic , Sterol Regulatory Element Binding Protein 1/metabolism , Cell Count , Cell Death , Cell Hypoxia , Cell Proliferation , Cell Survival/drug effects , DNA Fragmentation , Electrophoresis, Gel, Two-Dimensional , Fatty Acid Synthase, Type I/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glucose/pharmacology , Hep G2 Cells , Humans , Malonyl Coenzyme A/genetics , Malonyl Coenzyme A/metabolism , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 1/genetics , Transfection
12.
Oncol Res ; 19(10-11): 455-61, 2011.
Article in English | MEDLINE | ID: mdl-22715588

ABSTRACT

Coenzyme Q10 (CoQ10) is an essential factor of the mitochondrial respiratory chain and has effective antioxidant properties. Therefore, CoQ10 has been used in a variety of clinical applications and used as a nutritional supplement to treat several human diseases. Here, we tested the effects of CoQ10 on angiogenesis stimulated by basic fibroblast growth factor (bFGF). CoQ10 significantly inhibited bFGF-induced angiogenesis in a mouse Matrigel plug and the sprouting of endothelial cells in rat aortic rings. In addition, CoQ10 decreased the ability of tube formation, migration, and invasion in endothelial cells. When CoQ10 was used to inhibit angiogenesis in endothelial cells, the expression of vascular endothelial growth factor (VEGF) and the phosphorylation of ERK were decreased. Taken together, these results indicate that CoQ10 is able to act as an antiangiogenic regulator, and its inhibitory activity is mediated by blocking an ERK-dependent pathway. This study suggests that CoQ10 may be used a therapeutic agent to decrease neovascularization in several diseases, including solid tumors.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblast Growth Factor 2/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Ubiquinone/analogs & derivatives , Animals , Cells, Cultured , Enzyme Activation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Ubiquinone/pharmacology , Vascular Endothelial Growth Factor A/physiology
13.
Cell Death Differ ; 28(11): 3092-3104, 2021 11.
Article in English | MEDLINE | ID: mdl-34007068

ABSTRACT

The basic function of ß-arrestin 2 (Arrb2) is to negatively regulate the G-protein-coupled receptor signaling pathway through facilitating receptor desensitization and internalization. Arrb2 has also been reported to play various roles in cancer pathology including the proliferation, migration, invasion, metastasis, and apoptosis of solid tumors. However, the molecular mechanisms underlying the tumorigenic capacities of Arrb2 have not been elucidated. Here, we show a novel function of Arrb2: Arrb2 facilitates the degradation of HIF-1α, which is a master regulator of oxygen homeostasis. We also demonstrate that Arrb2 interacts with HIF-1α and stimulates ubiquitin-mediated 26S proteasomal degradation of HIF-1α by recruiting PHD2 and pVHL. Overexpression of Arrb2 in human glioblastoma cells suppresses HIF-1α signaling, tumor growth, and angiogenesis. Consistent with this antitumorigenic effect of Arrb2, low Arrb2 expression levels correlate with high HIF-1α expression and poor glioblastoma patient survival. These results collectively reveal a novel function of Arrb2 in the oxygen-sensing mechanism that directly regulates HIF-1α stability in human cancers and suggest Arrb2 as a new potential therapeutic target for glioblastoma.


Subject(s)
Glioblastoma/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , beta-Arrestin 2/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Rats , Transfection
14.
Oncol Lett ; 22(3): 671, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34345296

ABSTRACT

Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) have been used to treat patients with non-small cell lung cancer (NSCLC) and activating EGFR mutations; however, the emergence of secondary mutations in EGFR or the acquisition of resistance to EGFR-TKIs can develop and is involved in clinical failure. Since angiogenesis is associated with tumor progression and the blockade of antitumor drugs, inhibition of angiogenesis could be a rational strategy for developing anticancer drugs combined with EGFR-TKIs to treat patients with NSCLC. The signaling pathway mediated by hypoxia-inducible factor-1 (HIF-1) is essential for tumor angiogenesis. The present study aimed to identify the dependence of gefitinib resistance on HIF-1α activity using angiogenesis assays, western blot analysis, colony formation assay, xenograft tumor mouse model and immunohistochemical analysis of tumor tissues. In the NSCLC cell lines, HIF-1α protein expression levels and hypoxia-induced angiogenic activities were found to be increased. In a xenograft mouse tumor model, tumor tissues derived from gefitinib-resistant PC9 cells showed increased protein expression of HIF-1α and angiogenesis within the tumors. Furthermore, inhibition of HIF-1α suppressed resistance to gefitinib, whereas overexpression of HIF-1α increased resistance to gefitinib. The results from the present study provides evidence that HIF-1α was associated with the acquisition of resistance to gefitinib and suggested that inhibiting HIF-1α alleviated gefitinib resistance in NSCLC cell lines.

15.
Biochem Biophys Res Commun ; 391(1): 147-51, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19903456

ABSTRACT

The selective loss of dopaminergic neurons in the substantia nigra pars compacta is a feature of Parkinson's disease (PD). 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity is the most common experimental model used to investigate the pathogenesis of PD. Administration of MPTP in mice produces neuropathological defects as observed in PD and 1-methyl-4-pyridinium (MPP(+)) induces cell death when neuronal cell cultures are used. AMP-activated protein kinase (AMPK) is a key regulator of energy homeostasis. In the present study, we demonstrated that AMPK is activated by MPTP in mice and MPP(+) in SH-SY5Y cells. The inhibition of AMPK by compound C resulted in an increase in MPP(+)-induced cell death. We further showed that overexpression of AMPK increased cell viability after exposure to MPP(+) in SH-SY5Y cells. Based on these results, we suggest that activation of AMPK might prevent neuronal cell death and play a role as a survival factor in PD.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Neurons/enzymology , Parkinson Disease/enzymology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Apoptosis , Cell Line, Tumor , Dopamine Agents/pharmacology , Enzyme Activation , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects
16.
Biochem Biophys Res Commun ; 394(3): 854-8, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20307492

ABSTRACT

Ethyl pyruvate (EP), a simple derivative of endogenous pyruvate, has an anti-inflammatory function. Recently, the protective neurological effects of EP have been reported in cell culture and animal models of neurological diseases. The present study investigates the protective effects of EP on dopaminergic cell death in Parkinson's disease models. The selective death of dopaminergic neurons in substantia nigra was prevented by EP in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse models. EP also suppressed the 1-methyl-4-pyridinium-induced cell death of SH-SY5Y cells and restored the phosphorylation of extracellular signal-regulated kinase. Thus, EP has neuroprotective effects of EP in Parkinson's disease and its related signaling pathways.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Pyruvates/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Line , Disease Models, Animal , Dopamine/metabolism , Dopamine Agents/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Parkinson Disease/enzymology , Substantia Nigra/drug effects , Substantia Nigra/enzymology
17.
Cell Death Dis ; 11(8): 624, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796816

ABSTRACT

Although peripheral artery disease (PAD) is a major health problem, there have been limited advances in medical therapies. In PAD patients, angiogenesis is regarded as a promising therapeutic strategy to promote new arterial vessels and improve perfusion of ischemic tissue. Autophagy plays a critical role in catabolic processes for cell survival under normal and stressful conditions and plays fundamental biological roles in various cellular functions. In the present study, we showed that autophagy in endothelial cells is important for the repair and regeneration of damaged tissues. In a hindlimb ischemia mouse model, autophagy was stimulated in endothelial cells of the quadriceps muscle, and adjacent cells proliferated and regenerated. The autophagy pathway was induced under prolonged hypoxia in endothelial cells, and autophagy increased angiogenic activities. Moreover, conditioned media from endothelial cells blocked autophagy and inhibited the proliferation of muscle cells, suggesting that autophagic stimulation in endothelial cells affects the survival of adjacent cells, such as muscle. Collectively, hypoxia/ischemia-induced autophagy angiogenesis, and the damaged tissue surrounded by neo-vessels was regenerated in an ischemia model. Therefore, we strongly suggest that stimulation of autophagy in endothelial cells may be a potent therapeutic strategy in severe vascular diseases, including PAD.


Subject(s)
Autophagy , Endothelial Cells/pathology , Hindlimb/blood supply , Hindlimb/pathology , Ischemia/pathology , Neovascularization, Physiologic , Animals , Cell Hypoxia , Disease Models, Animal , Humans , Male , Mice, Inbred BALB C , Muscles/pathology , Rats, Sprague-Dawley , Stress, Physiological
18.
OMICS ; 22(7): 493-501, 2018 07.
Article in English | MEDLINE | ID: mdl-30004846

ABSTRACT

While progressive dopaminergic neurodegeneration is responsible for the cardinal motor defects in Parkinson's disease (PD), new diagnostics and therapeutic targets are necessary to effectively address this major global health burden. We evaluated whether the adhesion G protein-coupled receptor B1 (ADGRB1, formerly BAI1, brain-specific angiogenesis inhibitor 1) might contribute to dopaminergic neuronal loss. We used bioinformatic analyses, as well as in vitro and in vivo PD models. We report in this study that ADGRB1 is decreased in PD and that the ADGRB1 level is specifically decreased in dopaminergic neurons in the substantia nigra of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. In primary mouse mesencephalic neurons and human neuroblastoma cell lines, 1-methyl-4-phenylpyridinium (MPP+), a toxic metabolite of MPTP, suppressed the expression of ADGRB1. Moreover, we applied a network generation tool, Ingenuity Pathway Analysis®, with the transcriptomics dataset to extend the upstream regulatory pathway of ADGRB1 expression. AMP-activated protein kinase (AMPK) was predicted as a regulator, and consequently, 5-aminoimidazole-4-carboxamide ribonucleotide, a specific activator of AMPK, reduced the ADGRB1 protein level. Finally, ADGRB1 overexpression decreased nuclear condensation induced by MPP+ treatment. Taken together, we observed that decreased ADGRB1 by activation of AMPK induced neuronal cell death in MPTP/MPP+-mediated PD models, suggesting that ADGRB1 might potentially play a survival role in the neurodegenerative pathway of PD. These data offer new insights into dopaminergic cell death with therapeutic implications for neurodegenerative disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain/metabolism , Parkinson Disease/metabolism , Receptors, G-Protein-Coupled/metabolism , 1-Methyl-4-phenylpyridinium/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Blotting, Western , Cells, Cultured , Humans , Immunohistochemistry , Male , Mice , Parkinson Disease/genetics , Receptors, G-Protein-Coupled/genetics
19.
Biomed Opt Express ; 8(5): 2649-2659, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28663896

ABSTRACT

Non-thermal atmospheric-pressure plasma has been introduced in various applications such as sterilization, wound healing, blood coagulation, and other biomedical applications. The most attractive application of non-thermal atmospheric-pressure plasma is in cancer treatment, where the plasma is used to produce reactive oxygen species (ROS) to facilitate cell apoptosis. We investigate the effects of different durations of exposure to dielectric-barrier discharge (DBD) plasma on colon cancer cells using measurement of cell viability and ROS levels, western blot, immunocytochemistry, and Raman spectroscopy. Our results suggest that different kinds of plasma-treated cells can be differentiated from control cells using the Raman data.

20.
Physiol Res ; 65(6): 1031-1037, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27959575

ABSTRACT

Ginsenoside has been reported to have therapeutic effects for some types of cancer, but its effect on ovarian cancer cells has not been evaluated. In this study, we monitored the effects of ginsenoside-Rh2 (Rh2) on the inhibition of cell proliferation and the apoptotic process in the ovarian cancer cell line SKOV3 using an MTT assay and TUNEL assay. We found that Rh2 inhibited cell proliferation and significantly induced apoptosis. We confirmed the apoptotic effects of Rh2 using western blot analysis of apoptosis-related proteins. Specifically, the levels of cleaved poly ADP ribose polymerase (PARP) and cleaved caspase-3 significantly increased in SKOV3 cells treated with Rh2. Therefore, Rh2 clearly suppressed the growth of SKOV3 cells in vitro, which was associated with induction of the apoptosis pathway. Moreover, the migration assay showed that Rh2 inhibited the invasive ability of SKOV3 cells. Taken together, our results suggest that Rh2 has anticancer effects in SKOV3 cells through inhibition of cell proliferation and induction of apoptosis. Considering the therapeutic potential of Rh2, more studies should be carried out to facilitate the future application of this natural product as a potential anti-cancer agent.


Subject(s)
Caspase 3/metabolism , Ginsenosides/pharmacology , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis/drug effects , Caspase 3/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Proto-Oncogene Proteins c-bcl-2/drug effects , Signal Transduction/drug effects , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL