Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Biol Int ; 47(9): 1502-1518, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37208975

ABSTRACT

The transient receptor potential channel (TRP) channels are expressed in neuronal tissues and involved in neurological diseases such as pain, epilepsy, neuronal apoptosis, and neurodegenerative diseases. Formerly, we have investigated how neuronal differentiation changes TRP channels expression profile and how Parkinson's disease model is related with this expression levels. We have found that transient receptor potential channel melastatin subtype 7 (TRPM7), transient receptor potential channel melastatin subtype 8 and transient receptor potential channel vanilloid subtype 1 (TRPV1) channels have pivotal effects on differentiation and 1-Methyl-4-phenylpyridinium (MPP+ )-induced Parkinson's disease model in SH-SY5Y cells. In this study, we have investigated that downregulation of the TRP channels to evaluate how differentiation status changes to Parkinson's disease pathological hallmarks. We have also performed to other analyses to elucidate these TRP channels' function in MPP+ -induced neurotoxicity related apoptosis, cell viability, caspase 3 and 9 enzyme activities, intracellular reactive oxygen species production, mitochondrial depolarization levels, Ca2+ signaling, Alpha-synuclein and Dopamine levels, mono amino oxidase A and B enzymatic activities, both in differentiated and undifferentiated neuronal cells. Herein we have concluded that especially TRPM7 and TRPV1 channels have distinct role in Parkinson's disease pathology via their activity changings in pathological state, and downregulation of these channels or specific antagonists can be useful for the possible treatment strategy for Parkinson's disease and related markers.


Subject(s)
Neuroblastoma , Parkinson Disease , TRPM Cation Channels , Transient Receptor Potential Channels , Humans , Transient Receptor Potential Channels/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Down-Regulation , Apoptosis , 1-Methyl-4-phenylpyridinium/pharmacology , TRPV Cation Channels/metabolism , Protein Serine-Threonine Kinases/metabolism , Membrane Proteins/metabolism
2.
Metab Brain Dis ; 37(7): 2291-2304, 2022 10.
Article in English | MEDLINE | ID: mdl-35776390

ABSTRACT

Glia are essential neurons of the immune system in the central nervous system. The effective mission of glia depends on their activation, release of cytokines, and oxidative cleaning of debris material from neuronal cells. Accumulating evidence indicates that microglia activation-induced oxidative stress via the activation Ca2+ permeable TRPV1 channel has an essential role in the pathophysiology of neurodegenerative diseases. However, there is scarce information on the cytosolic localization of TRPV1 and the induction of oxidative cytotoxicity in the glia. Hence, we investigated the interactions between cytosolic TRPV1 expression levels and oxidative neurotoxicity in the BV2, C8-D1A, N9 glia, and DBTRG glioblastoma cells. We observed TRPV1 expression in the perinuclear area but not in the cell membrane in the BV2, C8-D1A, and N9 cells. Hence, we observed no activation of TRPV1 on the increase of mitochondrial free reactive oxygen species (mROS) and apoptosis in the cells after the capsaicin stimulation. However, we observed TRPV1 channel expression in the positive control (DBTRG) cell membranes. Hence, the Ca2+ influx, TRPV1 current density, apoptosis, and mROS levels were increased in the DBTRG cells after the capsaicin stimulation, although their levels were diminished by the treatment of the TRPV1 blocker (capsazepine). In conclusion, the presence of TRPV1 in the cell membrane of DBTRG cells induced excessive generation of mROS and apoptosis actions, although the presence of TRPV1 in the perinuclear area did not cause the actions. It seems that there is a subtype of TRPV1 in the perinuclear area, and it is not activated by the capsaicin.


Subject(s)
Capsaicin , TRPM Cation Channels , Capsaicin/pharmacology , Capsaicin/metabolism , TRPV Cation Channels/metabolism , Reactive Oxygen Species/metabolism , TRPM Cation Channels/metabolism , Ganglia, Spinal/metabolism , Hippocampus/metabolism , Calcium/metabolism , Apoptosis , Oxidative Stress , Neuroglia/metabolism , Cell Membrane/metabolism , Cytokines/metabolism
3.
Neurol Res ; 44(1): 24-37, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34256685

ABSTRACT

Transient Receptor Potential Melastatin-like 7 (TRPM7), Transient Receptor Potential Melastatin-like 8 (TRPM8) and Transient Receptor Potential Vanilloid-like 1 (TRPV1) channels are expressed in neurological tissues such as brain cortex, dorsal root ganglion and hippocampal neurons and involved in several neurological diseases. The SH-SY5Y neuronal cell line is frequently used as a cellular model of neurodegenerative diseases including Parkinson's disease. The differentiated SH-SY5Y cells have much neuronal structure, function and exaggerated neuronal marker expression. However, we have less data about how differentiation induces TRP channel expression and how TRP channels have a role in cellular functions in Parkinson's disease model in SH-SY5Y cells. Hence, we aimed to investigate the effects of differentiation phenomena on TRPM7, TRPM8 and TRPV1 cation channel expression and related Ca2+ signaling. We also made some other analysis to elucidate TRP channels' function in MPP induced apoptosis, mitochondrial membrane potential levels, intracellular reactive oxygen species production, caspase 3 and caspase 9 enzyme activities in differentiated or undifferentiated SH-SY5Y neuronal cells. Herein we concluded that TRPM7, TRPM8 and TRPV1 cation channels have pivotal effects on differentiation and MPP induced Parkinson's disease model in SH-SY5Y cells.


Subject(s)
Parkinson Disease , TRPM Cation Channels , Transient Receptor Potential Channels , Cell Differentiation , Ganglia, Spinal , Humans , Membrane Proteins/metabolism , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/pharmacology
4.
Curr Neuropharmacol ; 18(6): 501-517, 2020.
Article in English | MEDLINE | ID: mdl-31903884

ABSTRACT

Pain is a complex physiological process that includes many components. Growing evidence supports the idea that oxidative stress and Ca2+ signaling pathways participate in pain detection by neurons. The main source of endogenous reactive oxygen species (ROS) is mitochondrial dysfunction induced by membrane depolarization, which is in turn caused by Ca2+ influx into the cytosol of neurons. ROS are controlled by antioxidants, including selenium. Selenium plays an important role in the nervous system, including the brain, where it acts as a cofactor for glutathione peroxidase and is incorporated into selenoproteins involved in antioxidant defenses. It has neuroprotective effects through modulation of excessive ROS production, inflammation, and Ca2+ overload in several diseases, including inflammatory pain, hypersensitivity, allodynia, diabetic neuropathic pain, and nociceptive pain. Ca2+ entry across membranes is mediated by different channels, including transient receptor potential (TRP) channels, some of which (e.g., TRPA1, TRPM2, TRPV1, and TRPV4) can be activated by oxidative stress and have a role in the induction of peripheral pain. The results of recent studies indicate the modulator roles of selenium in peripheral pain through inhibition of TRP channels in the dorsal root ganglia of experimental animals. This review summarizes the protective role of selenium in TRP channel regulation, Ca2+ signaling, apoptosis, and mitochondrial oxidative stress in peripheral pain induction.


Subject(s)
Nervous System Diseases/physiopathology , Selenium/physiology , Animals , Calcium Signaling , Humans , Neuralgia/physiopathology , Transient Receptor Potential Channels/metabolism
5.
Int J Ophthalmol ; 12(6): 930-935, 2019.
Article in English | MEDLINE | ID: mdl-31236348

ABSTRACT

AIM: To observe the protective effect of astaxanthin (AST) against hydroquinone (HQ) mediated cell death in the apoptotic cascade and evaluate intracellular Ca2+ release, caspase-3, and -9 activation, reactive oxygen species (ROS) production in ARPE-19 cells. METHODS: We cultured ARPE-19 cells in special mediums and performed MTT tests to determine protective effect of AST, before exposing the cells to HQ in an incubator. We analyzed intracellular Ca2+ release experiments, mitochondrial membrane depolarization, glutathione (GSH), glutathione peroxidase (GSH-Px) and ROS experiments, and apoptosis assay. RESULTS: ROS production ranges depend on the amount of cell death. We computed the correlation between ROS ranges and cell death by 20,70-dichlorofluorescein fluorescence, and Ca2+ levels by Fura-2-AM. HQ-induced cell death found out to rise ranges of caspase-3 and -9, and mitochondrial depolarization. These three steps were delayed by AST management. CONCLUSION: ARPE-19 cells are avoided from HQ-induced ROS production and caspase-3 and -9 activation by AST. AST may limit the range of caspase synthesis, Ca2+ release and excess production of ROS with antiapoptotic effect. This study proposes a new therapeutic approach for the treatment of age-related macular degeneration.

6.
Biol Trace Elem Res ; 184(2): 358-368, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29081061

ABSTRACT

Increased intracellular free calcium ion (Ca2+) concentration induces excessive oxidative stress and apoptosis. Medical procedures such as zoledronic acid (Zol), bevacizumab (Bev), and dexamethasone (Dex) are usually used in the treatment of bone diseases (osteoporosis, Paget's disease, etc.) and to prevent metastasis in the bone although the procedures induce osteonecrosis of the jaw through excessive production of reactive oxygen species (ROS). Recently, we observed regulator roles of selenium (Se) on apoptosis and Ca2+ entry through transient receptor potential vanilloid 1 (TRPV1) channels in the cancer cell lines. Therefore, Se may modulate Zol, Bev, and Dex-induced oxidative stress and apoptosis through regulation of TRPV1 channel. In the current study, we investigated the protective effects of Se on apoptosis and oxidative stress through TRPV1 in Zol, Bev, and Dex-induced osteoblast-like cell line. We used human osteoblast-like cell line (Saos-2), and the cells were divided into 12 groups as control, Zol, Bev, Dex, Se, Zol+Se, Bev+Se, Dex+Se, Zol+Dex, Zol+Dex+Se, Zol+Bev, and Zol+Bev+Se which were incubated with drugs (Zol, Bev, Dex, and Se) for 24 h. The cytosolic free Ca2+ concentration was increased by Zol, Bev, Dex, Zol+Bev, and Zol+Dex, although it was reduced by Se treatment. However, Zol, Bev, and Dex-induced increase in apoptosis, caspase 3, caspase 9, poly (ADP-ribose) polymerase 1 expression levels, and intracellular ROS production values in the cells were decreased by Se treatments. In conclusion, we observed that Zol, Bev, and Dex-induced apoptosis, mitochondrial oxidative stress, and calcium signaling are decreased in human osteoblast-like cell line by the Se treatment. Our findings may be relevant to the etiology and treatment of Zol, Bev, and Dex-induced osteonecrosis by Se.


Subject(s)
Apoptosis/drug effects , Calcium/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Pharmaceutical Preparations/administration & dosage , Selenium/pharmacology , Bevacizumab/administration & dosage , Cell Line, Tumor , Dexamethasone/administration & dosage , Diphosphonates/administration & dosage , Humans , Imidazoles/administration & dosage , Mitochondria/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , TRPV Cation Channels/metabolism , Zoledronic Acid
7.
J Recept Signal Transduct Res ; 36(4): 395-401, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26608462

ABSTRACT

Neurological diseases such as Alzheimer's and Parkinson's diseases are incurable progressive neurological disorders caused by the degeneration of neuronal cells and characterized by motor and non-motor symptoms. Curcumin, a turmeric product, is an anti-inflammatory agent and an effective reactive oxygen and nitrogen species scavenging molecule. Hydrogen peroxide (H2O2) is the main source of oxidative stress, which is claimed to be the major source of neurological disorders. Hence, in this study we aimed to investigate the effect of curcumin on Ca(2+) signaling, oxidative stress parameters, mitochondrial depolarization levels and caspase-3 and -9 activities that are induced by the H2O2 model of oxidative stress in SH-SY5Y neuronal cells. SH-SY5Y neuronal cells were divided into four groups namely, the control, curcumin, H2O2, and curcumin + H2O2 groups. The dose and duration of curcumin and H2O2 were determined from published data. The cells in the curcumin, H2O2, and curcumin + H2O2 groups were incubated for 24 h with 5 µM curcumin and 100 µM H2O2. Lipid peroxidation and cytosolic free Ca(2+) concentrations were higher in the H2O2 group than in the control group; however, their levels were lower in the curcumin and curcumin + H2O2 groups than in the H2O2 group alone. Reduced glutathione (GSH) and glutathione peroxidase (GSH-Px) values were lower in the H2O2 group although they were higher in the curcumin and curcumin + H2O2 groups than in the H2O2 group. Caspase-3 activity was lower in the curcumin group than in the H2O2 group. In conclusion, curcumin strongly induced modulator effects on oxidative stress, intracellular Ca(2+) levels, and the caspase-3 and -9 values in an experimental oxidative stress model in SH-SY5Y cells.


Subject(s)
Apoptosis/drug effects , Calcium/metabolism , Curcumin/administration & dosage , Neurons/drug effects , Alzheimer Disease , Caspase 3/metabolism , Cell Polarity/drug effects , Cell Survival/drug effects , Humans , Hydrogen Peroxide/toxicity , Mitochondria/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Reactive Oxygen Species/metabolism , Supranuclear Palsy, Progressive/drug therapy , Supranuclear Palsy, Progressive/metabolism
8.
Mol Membr Biol ; 33(3-5): 76-88, 2016.
Article in English | MEDLINE | ID: mdl-28569571

ABSTRACT

Transient Receptor Potential (TRP) channels are mostly Ca2+ permeable cation channels. Transient Receptor Potential Melastatin-like 2 (TRPM2) is expressed in neurological tissues such as brain, dorsal root ganglia (DRG) neurons, hippocampus and also liver, heart and kidney. The SH-SY5Y cells are mostly used as a cellular model of neurodegenerative diseases, Alzheimer's and Parkinson's diseases. Curcumin, shows phenolic structure, synthesized by Curcuma longa L. (turmeric), has powerful non-enzymatically antioxidant effects compared with Vitamin E. Hence, we aimed to investigate that effects of curcumin on TRPM2 cation channel currents using the whole-cell Patch-Clamp method, Ca2+ signaling, apoptosis and cell viability (MTT) assays, reactive oxygen species (ROS) production, mitochondrial membrane potential levels, caspase 3 and caspase 9 activities in TRPM2 transfected SH-SY5Y neuroblastoma cells. For this aim, we designed four experimental groups named; control, curcumin, transfected and transfected + curcumin groups. Cytosolic free calcium concentrations were higher in transfected group compared with curcumin and transfected + curcumin group. Moreover, these data examined with whole-cell Patch-Clamp recordings of single cells in all groups. ROS levels were significantly higher in transfected group than in transfected + curcumin group. Apoptosis levels in transfected + curcumin group were lower than in transfected group. Procaspase 9 and procaspase 3 levels measured by western blotting and caspase 3 and caspase 9 levels by spectrophotometric methods show that TRPM2 transfected cells are more tended to apoptosis. In conclusion, curcumin strongly induces modulator effects on TRPM2-mediated Ca2+ influx caused by ROS and caspase 3 and 9 processes in SH-SY5Y neuroblastoma cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...