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








Publication year range
1.
Bioorg Med Chem Lett ; 108: 129816, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38806101

ABSTRACT

As our ongoing work, a novel series of the amide-based CA-4 analogues were successfully designed, synthesized, and explored for their biological evaluation. Among these compounds, 7d and 8a illustrated most potent antiproliferative activity toward A549, HeLa, HCT116, and HT-29 cell lines. Most importantly, these two compounds didn't display noticeable cytotoxic activity on the non-tumoural cell line HEK-293. Further mechanism studies revealed that analogue 8a was identified as a novel tubulin polymerization inhibitor with an IC50 value of 6.90 µM, which is comparable with CA-4. The subsequent investigations unveiled that analogue 8a not only effectively caused cell cycle arrest at the G2/M phase but also induced apoptosis in A549 cells via a concentration-dependent manner. The molecular docking revealed that 8a could occupy well the colchicine-binding site of tubulin. Collectively, these findings indicate that amide-based CA-4 scaffold could be worthy of further evaluation for development of novel tubulin inhibitors with improved safety profile.


Subject(s)
Amides , Antineoplastic Agents , Cell Proliferation , Drug Design , Drug Screening Assays, Antitumor , Molecular Docking Simulation , Stilbenes , Tubulin Modulators , Tubulin , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tubulin/metabolism , Structure-Activity Relationship , Amides/chemistry , Amides/pharmacology , Amides/chemical synthesis , Cell Proliferation/drug effects , Stilbenes/chemistry , Stilbenes/pharmacology , Stilbenes/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Molecular Structure , Cell Line, Tumor , Dose-Response Relationship, Drug , HEK293 Cells
2.
Eur J Pharmacol ; 899: 173908, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33515540

ABSTRACT

Rhein is one of the anthraquinones components of Rheum. It shows excellent clinical efficacy and is widely used in the management of several disease conditions including tumors, inflammation, diabetic nephropathy, and viral infections. In this review, we summarize the recent studies on the pharmacological activities of rhein and its derivatives, as well as their association with different diseases and possible mechanisms based on our previous review. This review serves as an updated and a supplement to our previous report highlighting the use of rhein in nanotechnology. It also serves as a reference study and offers an overall picture of the use of rhein and its derivatives in nanotechnology.


Subject(s)
Anthraquinones/therapeutic use , Animals , Anthraquinones/adverse effects , Anthraquinones/chemistry , Drug Carriers , Drug Compounding , Humans , Nanomedicine , Nanoparticles , Polymers/chemistry
3.
Eur J Pharmacol ; 883: 173361, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32673674

ABSTRACT

Alzheimer's disease (AD), which is characterized by impairment of cognitive functions, is a chronic neurodegenerative disease that mainly affects the elderly. Currently available anti-AD drugs can only offer limited symptom-relieving effects. "One-compound-Multitargeted Strategy" have been recognized as the promising way to win the war against AD. Herein we report a potential anti-AD agent PT109 with multi-functions. First, an 81-kinase screening was carried out and results showed that PT109 potently inhibited c-Jun N-terminal kinases and Serum and glucocorticoid-inducible kinase 1, which are the important signaling molecules involved in neurogenesis, neuroprotection and neuroinflammation and mildly inhibit glycogen synthase kinase-3ß as well as protein kinase C gamma, both are involved in AD pathological processes. In addition, invitro studies of immunofluorescent staining and Western blot showed that PT109 might promote the neurogenesis of C17.2 cells and induce synaptogenesis in primary cultured rat hippocampal neurons. We detected and confirmed the neuroprotective effect of PT109 in cultured HT22 cells by MTT assay, dehydrogenase assay, glutathione assay and reactive oxygen species assay. Furthermore, the results of Western blot, ELISA assay and immunofluorescent staining indicated that PT109 attenuated lipopolysaccharide-induced inflammation in BV2 cells and primary astrocytes. The results of Morris water maze and Step-through test indicated that PT109 improved the spatial learning ability in APP/PS1 mice. More importantly, the invivo pharmacokinetic parameters indicated that PT109 had better medicinal properties. Taken together, our findings suggest that PT109 may be a promising candidate for treating AD through multiple targets although further studies are ought to be conducted.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Drug Discovery , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Cell Line , Cytokines/metabolism , Disease Models, Animal , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Locomotion/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Molecular Targeted Therapy , Morris Water Maze Test/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacokinetics , Presenilin-1/genetics , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Signal Transduction
4.
Eur J Pharmacol ; 868: 172871, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31846627

ABSTRACT

Niacin has been widely used as an antihyperlipidemic drug, but the flushing effect restricted its clinical application. Here, we developed novel niacin-lipoic acid dimers which lead to better lipid modulation, higher synergistic effects and less side effects. We utilized molecular docking simulation to design a novel series of niacin-lipoic acid dimers. The compound N-(2-(5-(1,2-dithiolan-3-yl)pentanamido)ethyl)nicotinamide (N2L) was selected for the in vitro and in vivo evaluation, including the agonist activity in CHO-hGPR109A cells, cell protective effects in HT22 and HUVECs cells, flushing effect in guinea pigs and rats, lipid modulation in C57BL/6 mice and high fat diet-rats and atherosclerotic lesions regulation in apolipoprotein E null mice. N2L worked as potent and selective agonists for the high affinity niacin receptor GPR109A. N2L retained antioxidation and cytoprotection of lipoic acid. In addition, N2L displayed a good therapeutic index regarding lipid modulation and atherosclerotic lesions regulation, and minimized niacin-induced vasodilation (flushing) effect in vivo. N2L showed effective treatment regarding to lipid regulation and atherosclerosis inhibition effects, also with excellent antioxidant effects, safety profiles and non-flushing. All these results suggest N2L promising application prospects in the drug development for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Dyslipidemias/drug therapy , Flushing/prevention & control , Hypolipidemic Agents/pharmacology , Animals , Atherosclerosis/blood , Atherosclerosis/diagnosis , Cell Line , Cricetulus , Dimerization , Disease Models, Animal , Drug Design , Dyslipidemias/blood , Dyslipidemias/diagnosis , Female , Flushing/chemically induced , Human Umbilical Vein Endothelial Cells , Humans , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/therapeutic use , Lipids/blood , Male , Mice , Mice, Knockout, ApoE , Molecular Docking Simulation , Niacin/chemistry , Niacin/pharmacology , Niacin/therapeutic use , Rats , Thioctic Acid/chemistry , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use
5.
Invest New Drugs ; 38(3): 755-764, 2020 06.
Article in English | MEDLINE | ID: mdl-31414267

ABSTRACT

Glioblastoma multiforme (GBM) is the most common malignant tumor of the central nervous system (CNS). Effective treatments remain limited. Therefore, novel chemotherapy drugs with high efficiency and few adverse effects are urgently needed. Histone deacetylase (HDAC) and serum and glucocorticoid-regulated protein kinase 1 (SGK1) are targets for the prevention and treatment of GBM. Rhein has antitumor and SGK1 suppression effects, although its biological activity is limited by poor bioavailability. To improve the drug-like properties of rhein, we constructed a novel rhein-hydroxyethyl hydroxamic acid derivative (SYSUP007), which combined rhein with the HDAC inhibitor, suberoylanilide hydroxamic acid (SAHA). In the present study, the human GBM cell lines, T98G, U87 and U251, were used to investigate the anticancer effects of SYSUP007 in vitro. We found that SYSUP007 was more effective in inhibiting glioma cell proliferation, invasion and migration in vitro compared with the effects of rhein and SAHA. We also confirmed that SYSUP007 increased the expression of Ac-K100 and NDRG1 (targets of HDAC and SGK1). The present study indicates the potential that SYSUP007, as a novel rhein and SAHA derivative, for development as an anti-cancer therapy.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Biological Availability , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioblastoma/metabolism , Glioma/drug therapy , Glioma/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/metabolism , Immediate-Early Proteins/metabolism , Neoplasm Invasiveness/genetics , Protein Serine-Threonine Kinases/metabolism , Vorinostat/pharmacology
6.
Oncol Lett ; 13(3): 1990-1996, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28454354

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant type of primary brain tumor in adults and can diffusely infiltrate adjacent normal tissue. GBM is therefore rarely cured by surgery or radiation therapy. Matrix metalloproteinases (MMPs) are involved in tissue remodeling and numerous other physiological progresses. The MMPs MMP-2 and MMP-9 are associated with the invasion ability of GBM. PT93 is a novel caffeic acid amide derivative that was first synthesized in 2013. In the present study, the human GBM T98G, U87 and U251 cell lines and the normal mouse neuron HT22 cell line were used to investigate the anticancer and cytotoxic effects of PT93 in vitro. The cytotoxicity of PT93 was measured using MTT and lactate dehydrogenase assays. The anti-proliferation effect was tested using a cell colony formation assay. Gelatin zymography analysis and a scratch test were used to investigate the anti-migration mechanism of PT93. Western blot analysis was used to measure the expression of MMP-2/-9. The experimental results showed that PT93 suppressed the proliferation of T98G cells, and showed cytotoxicity effects at high concentration in T98G, U87, U251 and HT22 cell lines. Furthermore, PT93 limited the migration ability of the cells and inhibited the extracellular MMP-2 and MMP-9 activity of T98G and U251 cells. Finally, the present study confirmed that PT93 affects the level of MMP-2/-9 expression in T98G cells in a concentration-dependent manner. The present study indicates that PT93, as a novel caffeic acid amide derivative, may be used in the treatment of GBM.

7.
J Neuroimmunol ; 305: 108-114, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28284330

ABSTRACT

The microglia-mediated neuroinflammation plays an important role in the pathogenesis of Alzheimer's disease (AD). Advanced glycation end products (AGEs)/receptor for advanced glycation end products (RAGE) or Rho/Rho kinase (ROCK) are both involved in the development of non-specific inflammation. However, there are few reports about their effects on neuroinflammation. Here, we explored the mechanism of AGEs/RAGE/Rho/ROCK pathway underlying the non-specific inflammation and microglial polarization in BV2 cells. AGEs could activate ROCK pathway in a concentration-dependent manner. ROCK inhibitor fasudil and RAGE-specific blocker FPS-ZM1 significantly inhibited AGEs-mediated activation of BV2 cells and induction of reactive oxygen species (ROS). FPS-ZM1 and fasudil exerted their anti-inflammatory effects by downregulating inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), NLRP3 and nuclear translocation of nuclear factor kappa B (NF-κB) p65. In addition, AGEs induced both M1 (CD16/32, M1 marker) and M2 (CD206, M2 marker) phenotype in BV2 cells. Fasudil and FPS-ZM1 led to a decreased M1 and increased M2 phenotype. Together, these results indicate that the AGEs/RAGE/Rho/ROCK pathway in BV2 cells could intensify the non-specific inflammation of AD, which will provide novel strategies for the development of anti-AD drugs.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Polarity/physiology , Glycation End Products, Advanced/metabolism , Microfilament Proteins/metabolism , Microglia/physiology , NF-kappa B/metabolism , Signal Transduction/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Benzamides/pharmacology , Cardiac Myosins/metabolism , Cell Line, Transformed , Cell Polarity/drug effects , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/pharmacology , Mice , Microglia/classification , Microglia/drug effects , Myosin Light Chains/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , rho-Associated Kinases/metabolism
8.
Eur J Pharmacol ; 806: 1-9, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28320516

ABSTRACT

Microglia and astrocytes are largely responsible for inflammatory injury in the brain of Alzheimer's disease (AD). Increasing evidence has indicated that Rho kinase (ROCK) plays an important role in the regulation of neuroinflammation. Previously, we synthesized a new chemical entity L-F001 and proved its potential inhibitory effects on ROCK and oxidative stress. Here, we investigated the anti-inflammatory effects and the molecular mechanisms of L-F001 in vitro and in vivo. L-F001 remarkably suppressed lipopolysaccharides (LPS)-elevated expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) as well as LPS-induced production of nitric oxide (NO), reactive oxygen species, interleukin-6 (IL-6) and tumor necreactive oxygen speciesis factor-α (TNF-α) in microglial BV-2 cells and in cultured astrocytes. Furthermore, L-F001 inhibited the degradation of IκB and nuclear translocation of nuclear factor kappa B (NF-κB) p65 subunit. Moreover, L-F001 induced the upregulation of heme-oxygenase-1 (HO-1) and glutamate cysteine ligase modifier subunit (GCLM) expression, two downstream effectors of nuclear factor (erythroid-derived 2)-like 2 (Nrf2). It was interesting that L-F001 also activated phosphatidylinositol 3-kinase (PI3K) pathway and induced M1 (CD16/32, M1 marker)/ M2 (CD206, M2 maker) transition in BV-2 cells which was significantly blocked by a PI3K inhibitor, wortmannin. Finally, L-F001 markedly attenuated the level of pro-inflammatory mediators in a murine model of systemic acute brain inflammation induced by LPS. Taken together, these results indicate that the novel multifunctional ROCK inhibitor L-F001 suppresses neuroinflammation in vitro and in vivo via NF-κB inhibition and Nrf2 activation, suggesting that L-F001 may be a promising drug candidate for treating neuroinflammation-associated CNS diseases, including AD.


Subject(s)
Azepines/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Azepines/therapeutic use , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Female , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Sulfonamides/therapeutic use , Transcription Factor RelA/metabolism
9.
Neurochem Res ; 42(2): 615-624, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28078613

ABSTRACT

Amounting evidences demonstrated that Rho/Rho-associated kinase (ROCK) might be a novel target for the therapy of Parkinson's disease (PD). Recently, we synthesized L-F001 and revealed it was a potent ROCK inhibitor with multifunctional effects. Here we investigated the effects of L-F001 in PD models. We found that L-F001 potently attenuated 6-OHDA-induced cytotoxicity in PC12 cells and significantly decreased intracellular reactive oxygen species (ROS), prevented the 6-OHDA-induced decline of mitochondrial membrane potential and intracellular GSH levels. In addition, L-F001 increased Akt and GSK-3beta phosphorylation and induced the nuclear Nrf2 and HO-1 expression in a time- and concentration-dependent manner. Moreover, L-F001 restored the levels of p-Akt and p-GSK-3beta (Ser9) as well as HO-1 expression reduced by 6-OHDA. Those effects were blocked by the specific PI3K inhibitor, LY294002, indicating the involvement of Akt/GSK-3beta pathway in the neuroprotective effect of L-F001. In addition, L-F001 significantly attenuated the tyrosinehydroxylase immunoreactive cell loss in 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP)-induced mice PD model. Together, our findings suggest that L-F001 prevents 6-OHDA-induced cell death through activating Akt/GSK-3beta and Nrf2/HO-1 signaling pathway and attenuates MPTP-induced dopaminergic neuron toxicity in mice. L-F001 might be a promising drug candidate for PD.


Subject(s)
Azepines/pharmacology , Dopaminergic Neurons/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sulfonamides/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Death/drug effects , Cell Death/physiology , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , PC12 Cells , Protein Kinase Inhibitors/pharmacology , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , rho-Associated Kinases/antagonists & inhibitors
10.
Mol Neurobiol ; 54(3): 2209-2222, 2017 04.
Article in English | MEDLINE | ID: mdl-26941101

ABSTRACT

Methylene blue (MB) can ameliorate behavioral, neurochemical, and neuropathological impairments in animal models of acute and chronic neurodegenerative disorders, but the underlying mechanism remains unclear. Myocyte enhancer factor 2 (MEF2D) is known to promote neuronal survival in several models, and several survival and death signals converge on MEF2D and regulate its activity. Here, we investigated the role of MEF2D in the neuroprotective effect of MB against glutamate-induced toxicity in HT22 neuronal cells. Our results showed that MB, event at less than 100 nM, improved the viability of HT22 cells exposed to 2 mM glutamate. MB attenuated the mitochondrial impairment and quenches the reactive oxygen species (ROS) induced by glutamate. Surprisingly, MB at 50-200 nM did not affect the Nrf2/HO-1 pathway, an important endogenous anti-oxidative system. Further study showed that MB increased the transcription and translation of MEF2D. In addition, MB upregulated the expression of mitochondrial NADH dehydrogenase 6 (ND6) in a MEF2D-dependent manner. Knockdown of MEF2D abolished both MB-medicated increase of ND6 and MB-induced neuroprotection against glutamate-induced toxicity. Moreover, we showed that MB promoted Akt function activity, suppressed GSK-3ß activity, and increased MEF2D level in hippocampus of mice and HT22 cells. These findings for the first time demonstrate that MB protects HT22 neuronal cells against glutamate-induced cell death partially via the regulation of MEF2D-associated survival pathway.


Subject(s)
Hippocampus/drug effects , Methylene Blue/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hippocampus/metabolism , MEF2 Transcription Factors/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
11.
Eur J Med Chem ; 125: 784-792, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27736684

ABSTRACT

A series of rivastigmine-caffeic acid and rivastigmine-ferulic acid hybrids were designed, synthesized, and evaluated as multifunctional agents for Alzheimer's disease (AD) in vitro. The new compounds exerted antioxidant neuroprotective properties and good cholinesterases (ChE) inhibitory activities. Some of them also inhibited amyloid protein (Aß) aggregation. In particular, compound 5 emerged as promising drug candidates endowed with neuroprotective potential, ChE inhibitory, Aß self-aggregation inhibitory and copper chelation properties. These data suggest that compound 5 offers an attractive starting point for further lead optimization in the drug-discovery process against AD.


Subject(s)
Alzheimer Disease/drug therapy , Coumaric Acids/pharmacology , Rivastigmine/pharmacology , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Antioxidants/chemistry , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacokinetics , Coumaric Acids/chemistry , Drug Discovery/methods , Humans , In Vitro Techniques/methods , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Rivastigmine/chemistry , Structure-Activity Relationship
12.
Tumour Biol ; 37(11): 14721-14731, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27629139

ABSTRACT

The melastatin-like transient receptor potential 7 (TRPM7) has been implicated in proliferation or apoptosis of some cancers, indicating the potential of TRPM7 as an anti-anaplastic target. Here, we identified the characteristic TRPM7 channel currents in human malignant glioma MGR2 cells, which could be blocked by a pharmacologic inhibitor Gd3+. We mined the clinical sample data from Oncomine Database and found that human malignant glioma tissues expressed higher TRPM7 mRNA than normal brain ones. Importantly, we identified a widely used clinical anesthetic midazolam as a TRPM7 inhibitor. Midazolam treatment for seconds suppressed the TRPM7 currents and calcium influx, and treatment for 48 h inhibited the TRPM7 expression. The inhibitory effect on TRPM7 accounts for the proliferation loss and G0/G1 phase cell cycle arrest induced by midazolam. Our data demonstrates that midazolam represses proliferation of human malignant glioma cells through inhibiting TRPM7 currents, which may be further potentiated by suppressing the expression of TRPM7. Our result indicates midazolam as a pharmacologic lead compound with brain-blood barrier permeability for targeting TRPM7 in the glioma.


Subject(s)
Anti-Anxiety Agents/pharmacology , Calcium/metabolism , Cell Proliferation/drug effects , Glioma/drug therapy , Midazolam/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Data Mining , Databases, Factual , Fluorescent Antibody Technique , Glioma/metabolism , Glioma/pathology , Humans , Image Processing, Computer-Assisted/methods , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Tumor Cells, Cultured
13.
Neurochem Res ; 41(9): 2267-77, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27161377

ABSTRACT

Oxidative stress mediates the pathogenesis of neurodegenerative disorders. Gartanin, a natural xanthone of mangosteen, possesses multipharmacological activities. Herein, the neuroprotection capacity of gartanin against glutamate-induced damage in HT22 cells and its possible mechanism(s) were investigated for the first time. Glutamate resulted in cell death in a dose-dependent manner and supplementation of 1-10 µM gartanin prevented the detrimental effects of glutamate on cell survival. Additional investigations on the underlying mechanisms suggested that gartanin could effectively reduce glutamate-induced intracellular ROS generation and mitochondrial depolarization. We further found that gartanin induced HO-1 expression independent of nuclear factor erythroid-derived 2-like 2 (Nrf2). Subsequent studies revealed that the inhibitory effects of gartanin on glutamate-induced apoptosis were partially blocked by small interfering RNA-mediated knockdown of HO-1. Finally, the protein expression of phosphorylation of AMP-activated protein kinase (AMPK) and its downstream signal molecules, Sirtuin activator (SIRT1) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), increased after gartanin treatment. Taken together, these findings suggest gartanin is a potential neuroprotective agent against glutamate-induced oxidative injury partially through increasing Nrf-2-independed HO-1 and AMPK/SIRT1/PGC-1α signaling pathways.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Xanthones/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Glutamic Acid/pharmacology , Mice , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Xanthones/chemistry
14.
Neurochem Res ; 41(7): 1806-17, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27038926

ABSTRACT

Natural xanthones have diversity pharmacological activities. Here, a series of xanthones isolated from the pericarps of Garcinia mangostana Linn, named α-Mangostin, 8-Deoxygartanin, Gartanin, Garciniafuran, Garcinone C, Garcinone D, and γ-Mangostin were investigated. Biological screening performed in vitro and in Escherichia coli cells indicated that most of the xanthones exhibited significant inhibition of self-induced ß-amyloid (Aß) aggregation and also ß-site amyloid precursor protein-cleaving enzyme 1, acted as potential antioxidants and biometal chelators. Among these compounds, α-Mangostin, Gartanin, Garcinone C and γ-Mangostin showed better antioxidant properties to scavenge Diphenyl-1-(2,4,6-trinitrophenyl) hydrazyl (DPPH) free radical than Trolox, and potent neuroprotective effects against glutamate-induced HT22 cell death partly by up-regulating HO-1 protein level and then scavenging reactive oxygen species. Moreover, Gartanin, Garcinone C and γ-Mangostin could be able to penetrate the blood-brain barrier (BBB) in vitro. These findings suggest that the natural xanthones have multifunctional activities against Alzheimer's disease (AD) and could be promising compounds for the therapy of AD.


Subject(s)
Alzheimer Disease/metabolism , Garcinia mangostana , Plant Extracts/therapeutic use , Xanthones/therapeutic use , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Cell Line , Mice , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Xanthones/isolation & purification , Xanthones/pharmacology
15.
J Neurooncol ; 127(2): 261-70, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26725099

ABSTRACT

Survival of patients with glioblastoma (GBM) remains poor, and novel treatment methods are urgently needed. In this study, we tested the effects of a combination of fasudil, a ROCK inhibitor, and clioquinol, an 8-hydroxyquinoline derivative with antimicrobial properties, on human GBM U87 cells. Combination treatment synergistically inhibited the viability of glioma cells but not mouse normal neuron HT22 cells and significantly induced mitochondria-mediated apoptosis. Moreover, the combination was also found to trigger macro-autophagy (henceforth referred to as autophagy) by increasing the expression levels of several proteins involved in the induction of autophagy. Further studies showed that 3-methyladenine (3-MA) or chloroquine (CQ), two autophagy inhibitors, abrogated the cytotoxic effects of the combination treatment as well as the autophagy. Overall, we demonstrated that fasudil and clioquinol show synergistic anti-cancer effects, providing evidence for the further development of combination therapy for GBM.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Apoptosis/drug effects , Autophagy/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Clioquinol/pharmacology , Drug Synergism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Blotting, Western , Brain Neoplasms/metabolism , Cell Proliferation/drug effects , Drug Therapy, Combination , Flow Cytometry , Glioblastoma , Humans , Mice , Mitochondria/drug effects , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured
16.
Mol Cell Endocrinol ; 420: 169-79, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26499395

ABSTRACT

Neural stem cells (NSCs) are multipotent cells which are capable of self-replication and differentiation into neurons, astrocytes or oligodendrocytes in the central nervous system (CNS). NSCs are found in two main regions in the adult brain: the subgranular zone (SGZ) in the hippocampal dentate gyrus (DG) and the subventricular zone (SVZ). The recent discovery of NSCs in the adult mammalian brain has fostered a plethora of translational and preclinical studies to investigate novel approaches for the therapy of neurodegenerative diseases. Melatonin is the major secretory product synthesized and secreted by the pineal gland and shows both a wide distribution within phylogenetically distant organisms from bacteria to humans and a great functional versatility. Recently, accumulated experimental evidence showed that melatonin plays an important role in NSCs, including its proliferation, differentiation and survival, which are modulated by many factors including MAPK/ERK signaling pathway, histone acetylation, neurotrophic factors, transcription factors, and apoptotic genes. The purpose of this review is to summarize the beneficial effects of melatonin on NSCs and further to discuss the potential usage of melatonin and its derivatives or analogues in the treatment of CNS neurodegenerative diseases.


Subject(s)
Melatonin/analogs & derivatives , Melatonin/pharmacology , Neural Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Humans , Melatonin/chemistry , Models, Biological
17.
J Neurosci ; 34(34): 11426-38, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143622

ABSTRACT

Overstimulation of NMDA-type glutamate receptors is believed to be responsible for neuronal death of the CNS in various disorders, including cerebral and spinal cord ischemia. However, the intrinsic and physiological mechanisms of modulation of these receptors are essentially unknown. Here we report that cholestane-3ß,5α,6ß-triol (triol), a major metabolite of cholesterol, is an endogenous neuroprotectant and protects against neuronal injury both in vitro and in vivo via negative modulation of NMDA receptors. Treatment of cultured neurons with triol protects against glutamate-induced neurotoxicity, and administration of triol significantly decreases neuronal injury after spinal cord ischemia in rabbits and transient focal cerebral ischemia in rats. An inducible elevation of triol is associated with ischemic preconditioning and subsequent neuroprotection in the spinal cord of rabbits. This neuroprotection is effectively abolished by preadministration of a specific inhibitor of triol synthesis. Physiological concentrations of triol attenuate [Ca(2+)]i induced by glutamate and decrease inward NMDA-mediated currents in cultured cortical neurons and HEK-293 cells transiently transfected with NR1/NR2B NMDA receptors. Saturable binding of [(3)H]triol to cerebellar granule neurons and displacement of [(3)H]MK-801 binding to NMDA receptors by triol suggest that direct blockade of NMDA receptors may underlie the neuroprotective properties. Our findings suggest that the naturally occurring oxysterol, the major cholesterol metabolite triol, functions as an endogenous neuroprotectant in vivo, which may provide novel insights into understanding and developing potential therapeutics for disorders in the CNS.


Subject(s)
Brain Injuries/prevention & control , Cholestanols/therapeutic use , Neuroprotective Agents/therapeutic use , Spinal Cord Ischemia/prevention & control , Adult , Animals , Brain Injuries/etiology , Cells, Cultured , Central Nervous System/cytology , Cholestanols/blood , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Female , Glutamic Acid/pharmacology , Humans , Infarction, Middle Cerebral Artery/complications , Male , Neurons/drug effects , Neurons/physiology , Protein Binding/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution/drug effects , Tissue Distribution/physiology , Young Adult
18.
J Neurosurg Anesthesiol ; 25(3): 285-91, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23519373

ABSTRACT

BACKGROUND: Glioblastoma (GBM), the most common primary brain tumor, is the most aggressive malignancy in humans. Its rapid proliferation is a major obstacle to successful treatment. Patients with GBM often suffer from psychological disturbances associated with poor prognosis and physical discomfort. Diazepam is one of the most frequently used benzodiazepines (BZs) in cancer patients for its desirable psychotropic effects. The central effects of BZs are mediated by the activation of central BZ receptors. This study investigates whether diazepam has inhibitory effect on proliferation of GBM cell line T98G and explores its possible mechanism. METHODS: Cell viability and proliferation were respectively determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay and 5-bromo-2'-deoxyuridine (BrdU) incorporation assay. Cell cycle distribution was examined by flow cytometry. Western blot with specific protein antibodies was used to detect regulatory proteins involved in cell cycle regulation. RESULTS: Diazepam significantly decreased the proliferation of T98G cells in a dose-dependent and time-dependent manner. This effect was not reversed by the central BZ receptor antagonist flumazenil or the peripheral BZ receptor antagonist PK11195, indicating that it was not mediated by BZ receptors. Flow cytometry indicated that diazepam caused a cell accumulation in G0/G1 phase, thereby contributing to cell proliferation inhibition. Furthermore, our findings showed that lessened phosphorylation of Rb accounted for diazepam-induced G0/G1 phase arrest. CONCLUSIONS: Diazepam inhibits the proliferation of human GBM T98G cells by inducing G0/G1 phase arrest. Diazepam has potential to be a lead for new drugs in GBM therapy because of its antitumor activity.


Subject(s)
Brain Neoplasms/pathology , Cell Proliferation/drug effects , Diazepam/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , GABA Modulators/pharmacology , Glioblastoma/pathology , Antimetabolites , Benzimidazoles , Blotting, Western , Bromodeoxyuridine , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Coloring Agents , Fluorescent Dyes , G1 Phase/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Resting Phase, Cell Cycle/drug effects , S Phase/drug effects , Tetrazolium Salts , Thiazoles
19.
Oncol Lett ; 5(3): 1010-1016, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23426784

ABSTRACT

Transient receptor potential melastatin 7 (TRPM7), a Ca(2+)-permeable channel, has been demonstrated to be present in cancer cells and involved in their growth and proliferation. The present study used midazolam, a benzodiazepine class anesthesic, to pharmacologically intervene in the expression of TRPM7 and to inhibit cancer cell proliferation. Midazolam significantly inhibited the growth and proliferation of FaDu human hypopharyngeal squamous cell carcinoma cells, concurring with the induction of G(0)/G(1) cell cycle arrest and blockage of Rb activation. Central-type and peripheral-type benzodiazepine receptor antagonists did not abrogate proliferation inhibition by midazolam, while the specific TRPM7 agonist bradykinin reversed this effect. In addition, other benzodiazepines, diazepam and clonazepam also exhibited anti-proliferative activities. The inhibitory activity on cancer cell growth and proliferation, combined with the TRPM-dependent mechanism, reveals the anticancer potential of midazolam as a TRPM7 inhibitor and supports the suggestion that TRPM7 is a valuable target for pharmaceutical intervention.

20.
Oncol Rep ; 29(2): 541-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23165370

ABSTRACT

Tumor invasion and migration are major causes of mortality in patients with cervical carcinoma. Tumors under hypoxic conditions are more invasive and have a higher metastasic activity. Lysyl oxidase (LOX) is a hypoxia-responsive gene. LOX has been shown to be essential for hypoxia-induced metastasis in breast cancer. However, the direct impact of LOX on cervical cancer cell motility remains poorly understood. Our study revealed that LOX expression at protein and catalytic levels is upregulated in cervical cancer cells upon exposure to hypoxia. Hypoxia induced mesenchymal-like morphological changes in HeLa and SiHa cells which were accompanied by upregulation of α-SMA and vimentin, two mesenchymal markers, and downregulation of E-cadherin, an epithelial marker, indicating the epithelial-mesenchymal transition (EMT) of cervical cancer cells occurred under hypoxic conditions. Treatment of tumor cells with ß-aminopropionitrile (BAPN), an active site inhibitor of LOX, blocked the hypoxia-induced EMT morphological and marker protein changes, and inhibited invasion and migration capacities of cervical carcinoma cells in vitro. Collectively, these findings suggest LOX enhances hypoxia-induced invasion and migration in cervical cancer cells mediated by the EMT which can be inhibited by BAPN.


Subject(s)
Aminopropionitrile/pharmacology , Cell Movement/physiology , Enzyme Inhibitors/pharmacology , Epithelial-Mesenchymal Transition/physiology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/pathology , Actins/metabolism , Cadherins/metabolism , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , HeLa Cells , Humans , Hypoxia/physiopathology , Neoplasm Invasiveness , Protein-Lysine 6-Oxidase/metabolism , Up-Regulation , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL