Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Anat Sci Int ; 98(4): 529-539, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37024641

ABSTRACT

Traumatic axonal damage disrupts connections between neurons, leading to the loss of motor and sensory functions. Although damaged peripheral nerves can regenerate, recovery depends on the variety and severity of nerve damage. Thus, many phytochemicals have been studied for their ability to reduce peripheral nerve degeneration, and among them, Parthenolide (PTL), which is extracted from Feverfew has effects against production of free radicals, inflammation, and apoptosis. Thus, we conducted a study to investigate whether PTL has an inhibitory effect on peripheral nerve degeneration during peripheral nerve damage. To verify the effect of PTL on peripheral nerve degeneration process, a morphological comparison of peripheral nerves with and without PTL was performed. PTL significantly reduced the quantity of fragmented ovoid formations at 3DIV (days in vitro). Immunostaining for MBP revealed that the ratio of intact myelin sheaths increased significantly in sciatic nerve with PTL compared with absence of PTL at 3DIV. Furthermore, nerve fibers in the presence of PTL maintained the continuity of Neurofilament (NF) compared to those without at 3DIV. Immunostaining for LAMP1 and p75 NTR showed that the expression of LAMP1 and p75 NTR decreased in the nerve after PTL addition at 3DIV. Lastly, immunostaining for anti-Ki67 revealed that PTL inhibited Ki67 expression at 3DIV compared to without PTL. These results confirm that PTL inhibits peripheral nerve degenerative processes. PTL may be a good applicant to inhibit peripheral nerve degeneration. Our study examined the effect of Parthenolide in preventing degeneration of peripheral nerves by inhibiting the breakdown of peripheral axons and myelin, also inhibiting Schwann cell trans-dedifferentiation and proliferation.


Subject(s)
Peripheral Nervous System Diseases , Sesquiterpenes , Humans , Axons , Schwann Cells/pathology , Sesquiterpenes/pharmacology , Sesquiterpenes/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Sciatic Nerve/pathology , Peripheral Nervous System Diseases/pathology , Nerve Regeneration/physiology
2.
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
3.
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.

4.
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
5.
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
6.
Chem Sci ; 10(24): 6157-6161, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31360422

ABSTRACT

Shape control of metal-organic materials on the meso- and macroscale has been an important theme due to emerging properties. Particularly, chemical etching has been useful to create various forms such as core-shells and hollow crystals in metal-organic frameworks. Here we present a unique chemical etching strategy to create trigonal patterned surfaces in metal-organic frameworks. The mechanism suggests that metal-organic polyhedron subunits serve as meta-atoms, playing a crucial role in the formation of trigons on the surface. Such a patterned surface in porous solids can be utilized in meta-surface applications in the foreseeable future.

7.
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
8.
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
9.
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
10.
Inorg Chem ; 57(9): 5225-5231, 2018 May 07.
Article in English | MEDLINE | ID: mdl-29664295

ABSTRACT

Open coordination sites (OCSs) in metal-organic frameworks (MOFs) have shown potential in applications such as molecular separation, sorption, catalysis, and sensing. Thus, the removal of coordinated solvent has been viewed as an essential step that needs to be performed prior to the use of the MOFs in the above applications. To date, a thermal method that is normally performed by applying heat and vacuum has been the most commonly employed activation method despite its negative influence on the structural integrity of the MOFs. In this report, we demonstrate that commonly inert trichloromethane (TCM) can activate OCSs; the TCM treatment process serves as an alternative chemical route to activation that does not require the external thermal energy. On the basis of the Raman study, we suggest a possible mechanism for the chemical activation process where TCM may weakly coordinate to the OCSs and then spontaneously dissociate. In addition, we prove that the chemical activation behavior is substantially boosted when a small amount of external heat energy (55 °C, 2.6 meV) is supplied during the TCM treatment. Using an HKUST-1-polyvinylidene fluoride (PVDF) mixed matrix (MM), we also demonstrate that this chemical activation strategy is a safe way to activate thermally deformable MOF-polymer mixed matrices.

11.
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
12.
ACS Appl Mater Interfaces ; 10(4): 3793-3800, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-29297676

ABSTRACT

Ionic polymers that possess ion-exchangeable sites have been shown to be a greatly useful platform to fabricate mixed matrices (MMs) where metal-organic frameworks (MOFs) can be in situ synthesized, although the in situ synthesis of MOF has been rarely studied. In this study, alginate (ALG), an anionic green polymer that possesses metal-ion-exchangeable sites, is employed as a platform of MMs for the in situ synthesis of iconic MOFs, HKUST-1, and MOF-74(Zn). We demonstrate for the first time that the sequential order of supplying MOF ingredients (metal ion and deprotonated ligand) into the alginate matrix leads to substantially different results because of a difference in the diffusion of the MOF components. For the examples examined, whereas the infusion of BTC3- ligand into Cu2+-exchanged ALG engendered the eggshell-shaped HKUST-1 layers on the surface of MM spheres, the infusion of Cu2+ ions into BTC3--included alginate engendered the high dispersivity and junction contact of HKUST-1 crystals in the alginate matrix. This fundamental property has been exploited to fabricate a flexible MOF-containing mixed matrix membrane by coincorporating poly(vinyl alcohol). Using two molecular dyes, namely, methylene blue and rhodamine 6G, further, we show that this in situ strategy is suitable for fabricating an MOF-MM that exhibits size-selective molecular uptake.

13.
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.

14.
ACS Appl Mater Interfaces ; 9(29): 24743-24752, 2017 Jul 26.
Article in English | MEDLINE | ID: mdl-28671454

ABSTRACT

The activation of open coordination sites (OCSs) in metal-organic frameworks (MOFs), i.e., the removal of solvent molecules coordinated at the OCSs, is an essential step that is required prior to the use of MOFs in potential applications such as gas chemisorption, separation, and catalysis because OCSs often serve as key sites in these applications. Recently, we developed a "chemical activation" method involving dichloromethane (DCM) treatment at room temperature, which is considered to be a promising alternative to conventional thermal activation (TA), because it does not require the application of external thermal energy, thereby preserving the structural integrity of the MOFs. However, strongly coordinating solvents such as N,N-dimethylformamide (DMF), N,N-diethylformamide (DEF), and dimethyl sulfoxide (DMSO) are difficult to remove solely with the DCM treatment. In this report, we demonstrate a multiple coordination exchange (CE) process executed initially with acetonitrile (MeCN), methanol (MeOH), or ethanol (EtOH) and subsequently with DCM to achieve the complete activation of OCSs that possess strong extracoordination. Thus, this process can serve as an effective "chemical route" to activation at room temperature that does not require applying heat. To the best of our knowledge, no previous study has demonstrated the activation of OCSs using this multiple CE process, although MeOH and/or DCM has been popularly used in pretreatment steps prior to the TA process. Using MOF-74(Ni), we demonstrate that this multiple CE process can safely activate a thermally unstable MOF without inflicting structural damage. Furthermore, on the basis of in situ 1H nuclear magnetic resonance (1H NMR) and Raman studies, we propose a plausible mechanism for the activation behavior of multiple CE.

15.
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
16.
Oncotarget ; 7(30): 47232-47241, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27323807

ABSTRACT

Angiogenesis is an essential step for tumor survival and progression, and the inhibition of angiogenesis is a good strategy for tumor therapeutics. In this study, we investigated the therapeutic effect of zingerone in a mouse tumor model. Zingerone suppressed tumor progression and tumor angiogenesis. Moreover, we found that zingerone inhibited the angiogenic activities of endothelial cells by both direct and indirect means. A mechanistic study showed that the activities of MMP-2 and MMP-9 in tumor cells were decreased by treatment with zingerone. Interestingly, zingerone-mediated inhibition of MMP-2 and MMP-9 was involved in the JNK pathway. In conclusion, zingerone showed strong anti-angiogenic activity via the inhibition of MMP-2 and MMP-9 during tumor progression, suggesting that zingerone may be a potential therapeutic drug for human cancers.


Subject(s)
Guaiacol/analogs & derivatives , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Matrix Metalloproteinase Inhibitors/pharmacology , Animals , Cell Growth Processes/drug effects , Disease Models, Animal , Guaiacol/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/enzymology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology
17.
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
18.
Biochem Pharmacol ; 98(1): 41-50, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26297910

ABSTRACT

During tumor progression, hypoxia-inducible factor 1 (HIF-1) plays a critical role in tumor angiogenesis and tumor growth by regulating the transcription of several genes in response to a hypoxic environment and changes in growth factors. This study was designed to investigate the effects of cinnamic aldehyde (CA) on tumor growth and angiogenesis and the mechanisms underlying CA's anti-angiogenic activities. We found that CA administration inhibits tumor growth and blocks tumor angiogenesis in BALB/c mice. In addition, CA treatment decreased HIF-1α protein expression and vascular endothelial growth factor (VEGF) expression in mouse tumors and Renca cells exposed to hypoxia in vitro. Interestingly, CA treatment did not affect the stability of von Hippel-Lindau protein (pVHL)-associated HIF-1α and CA attenuated the activation of mammalian target of rapamycin (mTOR) pathway. Collectively, these findings strongly indicate that the anti-angiogenic activity of CA is, at least in part, regulated by the mTOR pathway-mediated suppression of HIF-1α protein expression and these findings suggest that CA may be a potential drug for human cancer therapy.


Subject(s)
Acrolein/analogs & derivatives , Antineoplastic Agents, Phytogenic/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia/metabolism , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/prevention & control , Acrolein/pharmacology , Animals , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...