Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 236
Filter
Add more filters










Publication year range
1.
Clin Exp Pharmacol Physiol ; 51(4): e13844, 2024 04.
Article in English | MEDLINE | ID: mdl-38350599

ABSTRACT

Botulinum neurotoxin A (BoNT) is being shown to have anticancer action as a potential adjuvant treatment. The transient receptor potential (TRP) melastatin 2 (TRPM2) stimulator action of BoNT was reported in glioblastoma cells, but not in colorectal cancer (HT29) cells. By activating TRPM2, we evaluated the impacts of BoNT and oxaliplatin (OXA) incubations on oxidant and apoptotic values within the HT29 cells. Control, BoNT (5 IU for 24 h), OXA (50 µM for 24 h) and their combinations were induced. We found that TRPM2 protein is upregulated and mediates enhanced BoNT and OXA-induced Ca2+ entry in cells as compared to control cells. The increase of free reactive oxygen species (ROS), but the decrease of glutathione is the main ROS responsible for TRPM2 activation on H29 exposure to oxidative stress. BoNT and OXA-mediated Ca2+ entry through TRPM2 stimulation in response to H2 O2 results in mitochondrial Ca2+ overload, followed by mitochondrial membrane depolarization, apoptosis and caspase-3/-8/-9, although they were diminished in the TRPM2 antagonist groups (N-(p-amylcinnamoyl)anthranilic acid and carvacrol). In conclusion, by increasing the susceptibility of HT29 tumour cells to oxidative stress and apoptosis, the combined administration of BoNT and OXA via the targeting of TRPM2 may offer a different approach to kill the tumour cells.


Subject(s)
Botulinum Toxins, Type A , Colorectal Neoplasms , TRPM Cation Channels , Humans , Oxaliplatin/pharmacology , Reactive Oxygen Species/metabolism , Botulinum Toxins, Type A/metabolism , Up-Regulation , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Cell Death , Oxidative Stress/physiology , Apoptosis/physiology , Colorectal Neoplasms/drug therapy , Calcium/metabolism
3.
J Bioenerg Biomembr ; 55(3): 179-193, 2023 06.
Article in English | MEDLINE | ID: mdl-37357235

ABSTRACT

Diabetes mellitus (DM) is a chronic syndrome involving neuropathic pain. Increased oxidative stress in DM is assumed to increase free reactive oxygen radicals (ROS) and causes diabetic damage. The sciatic nerve (ScN) and dorsal root ganglion (DRG) both contain high levels of the TRPV1 channel, which is triggered by capsaicin and ROSs and results in increased Ca2+ entry into the neurons. Alpha-lipoic acid (ALA) is considered an important part of the antioxidant system. To better characterize the protective effects of ALA on the DM-induced neuronal through TRPV1 modulation, we investigated the role of ALA on DM-induced neuropathic pain, oxidative ScN, and DRG damage in diabetic rats. Forty adult Wistar albino female rats were divided into four groups as control, ALA (50 mg/kg for 14 days), streptozotocin (STZ and 45 mg/kg and single dose), and STZ + ALA. Rats were used for the pain tests. After obtaining the DRGs and ScN, they were used for plate reader, patch-clamp, and laser confocal microscope analyses. We observed the modulator role of ALA on the thresholds of mechanical withdrawal pain (von Frey test) and hot sensitivity pain (hot plate test) in the STZ + ALA group. The treatment of ALA decreased STZ-induced increase of TRPV1 current densities, intracellular free Ca2+ concentrations (Fura-2 and Fluo - 3/AM), ROS, caspase 3, caspase 9, mitochondrial membrane potential, and apoptosis values in the ScN and DRG neurons, although its treatment induced the increase of cell viability and body weight gain. The treatment of ALA acted a neuroprotective role on the TRPV1 channel stimulation-mediated Ca2+ influx, neuropathic pain, and neuronal damage in diabetic rats. The neuroprotective role of ALA treatment can be explained by its modulating the TRPV1 channel activity, intracellular Ca2+ increase-induced oxidative stress, and apoptosis.


Subject(s)
Diabetes Mellitus, Experimental , Neuralgia , Thioctic Acid , Rats , Animals , Rats, Wistar , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use , Reactive Oxygen Species/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Oxidative Stress , Apoptosis , Neuralgia/drug therapy , Ganglia, Spinal/metabolism , TRPV Cation Channels/metabolism , TRPV Cation Channels/pharmacology
4.
Graefes Arch Clin Exp Ophthalmol ; 261(10): 2829-2844, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37099129

ABSTRACT

PURPOSE: Hydroxychloroquine (HCQ) is used in the treatment of several diseases, such as malaria, Sjögren's disease, Covid-19, and rheumatoid arthritis. However, HCQ induces retinal pigment epithelium death via the excessive increase of cytosolic (cROS) and mitochondrial (mROS) free oxygen radical production. The transient receptor potential melastatin 2 (TRPM2) cation channel is stimulated by ADP-ribose (ADPR), cROS, and mROS, although it is inhibited by curcumin (CRC). We aimed to investigate the modulating action of CRC on HCQ-induced TRPM2 stimulation, cROS, mROS, apoptosis, and death in an adult retinal pigment epithelial 19 (ARPE19) cell line model. MATERIAL AND METHODS: ARPE19 cells were divided into four groups: control (CNT), CRC (5 µM for 24 h), HCQ (60 µM for 48 h), and CRC + HCQ groups. RESULTS: The levels of cell death (propidium iodide positive cell numbers), apoptosis markers (caspases -3, -8, and -9), oxidative stress (cROS and mROS), mitochondria membrane depolarization, TRPM2 current density, and intracellular free Ca2+ and Zn2+ fluorescence intensity were upregulated in the HCQ group after stimulation with hydrogen peroxide and ADPR, but their levels were downregulated by treatments with CRC and TRPM2 blockers (ACA and carvacrol). The HCQ-induced decrease in retinal live cell count and cell viability was counteracted by treatment with CRC. CONCLUSION: HCQ-mediated overload Ca2+ influx and retinal oxidative toxicity were induced in an ARPE19 cell line through the stimulation of TRPM2, although they were attenuated by treatment with CRC. Hence, CRC may be a potential therapeutic antioxidant for TRPM2 activation and HCQ treatment-induced retinal oxidative injury and apoptosis.


Subject(s)
COVID-19 , Curcumin , TRPM Cation Channels , Humans , Adenosine Diphosphate Ribose/metabolism , Apoptosis , Calcium , Cell Line , COVID-19 Drug Treatment , Curcumin/pharmacology , Hydroxychloroquine/pharmacology , Oxidative Stress/physiology , Retinal Pigment Epithelium/metabolism , TRPM Cation Channels/metabolism
5.
J Obstet Gynaecol Res ; 49(5): 1355-1365, 2023 May.
Article in English | MEDLINE | ID: mdl-36807656

ABSTRACT

AIM: Recurrent pregnancy loss (RPL) is known to be associated with increased thrombophilia and oxidative toxicity. However, the mechanism of thrombophilia apoptosis and oxidative toxicity is still unclear. In addition, the treatment of heparin induced regulator roles on intracellular free Ca2+ ([Ca2+ ]i ) and cytosolic reactive oxygen species (cytROS) concentrations in several diseases. TRPM2 and TRPV1 channels are activated by different stimuli, including oxidative toxicity. The aim of this study was to investigate the effects of low molecular weight heparin (LMWH) via modulation of TRPM2 and TRPV1 on calcium signaling, oxidative toxicity, and apoptosis in the thrombocytes of RPL patients. STUDY DESIGN: Thrombocyte and plasma samples collected from 10 patients with RPL and 10 healthy controls were used in the current study. MAIN FINDINGS: The [Ca2+ ]i concentration, cytROS (DCFH-DA), mitochondrial membrane potential (JC-1), apoptosis, caspase-3, and caspase-9 levels were high in the plasma and thrombocytes of RPL patients, although they were diminished by the treatments of LMWH, TRPM2 (N-(p-amylcinnamoyl)anthranilic acid) and TRPV1 (capsazepine) channel blockers. CONCLUSIONS: The current study results suggest that the treatment of LMWH is useful against apoptotic cell death and oxidative toxicity in the thrombocytes of patients with RPL, which seem to be dependent on increased levels of [Ca2+ ]i concentration via the activation of TRPM2 and TRPV1.


Subject(s)
Oxidative Stress , TRPM Cation Channels , Rats , Animals , Humans , TRPM Cation Channels/metabolism , Blood Platelets/metabolism , Rats, Wistar , Heparin, Low-Molecular-Weight/pharmacology , Reactive Oxygen Species/metabolism , Apoptosis , Calcium/metabolism , Calcium/pharmacology , TRPV Cation Channels/metabolism , TRPV Cation Channels/pharmacology
6.
Brain Res ; 1803: 148232, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36610553

ABSTRACT

TRPM2 channel is activated by the increase of hypoxia (HYP)-mediated excessive mitochondrial (mROS) and cytosolic (cROS) free reactive oxygen species generation and intracellular free Ca2+ ([Ca2+]i) influx. The stimulations of the N-methyl-d-aspartate(NMDA) receptor and TRPM2 channel induce mROS and apoptosis in the neurons, although their inhibitions via the treatments of memantine (MEM) and MK-801 decrease mROS and apoptosis. However, the molecular mechanisms underlying MEM treatment and NMDA inhibition' neuroprotection via TRPM2 inhibition in the HYP remain elusive. We investigated the modulator role of MEM and NMDA via the modulation of TRPM2 on oxidative neurodegeneration and apoptosis in SH-SY5Y neuronal cells. Six groups were induced in the SH-SY5Y and HEK293 cells as follows: Control, MEM, NMDA blocker (MK-801), HYP (CoCl2), HYP + MEM, and HYP + MK-801. The HYP caused to the increases of TRPM2 and PARP-1 expressions, and TRPM2 agonist (H2O2 and ADP-ribose)-induced TRPM2 current density and [Ca2+]i concentration via the upregulation of mitochondrial membrane potential, cROS, and mROS generations. The alterations were not observed in the absence of TRPM2 in the HEK293 cells. The increase of cROS, mROS, lipid peroxidation, cell death (propidium iodide/Hoechst) rate, apoptosis, caspase -3, caspase -8, and caspase -9 were restored via upregulation of glutathione and glutathione peroxidase by the treatments of TRPM2 antagonists (ACA or 2-APB), MEM, and MK-801. In conclusion, the inhibition of NMDA receptor via MEM treatment modulated HYP-mediated mROS, apoptosis, and TRPM2-induced excessive [Ca2+]i and may provide an avenue for protecting HYP-mediated neurodegenerative diseases associated with the increase of mROS, [Ca2+]i, and apoptosis.


Subject(s)
Neuroblastoma , TRPM Cation Channels , Humans , Receptors, N-Methyl-D-Aspartate/metabolism , Memantine/pharmacology , TRPM Cation Channels/metabolism , Oxidative Stress/physiology , Dizocilpine Maleate/pharmacology , Hydrogen Peroxide/metabolism , HEK293 Cells , N-Methylaspartate/metabolism , Apoptosis/physiology , Hypoxia , Neurons/metabolism , Calcium/metabolism , Reactive Oxygen Species/metabolism
7.
Toxicon ; 222: 106993, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36528210

ABSTRACT

Melittin (MLT) treatment is believed to enhance tumor cell death, apoptotic, and oxidative cytotoxic effects of cisplatin (CSP) via the modulation of Ca2+ channels in several cancer lines. The activation of TRPM2 mediated anticancer and CSP resistance actions via mitochondrial Ca2+ and Zn2+ accumulation-induced mitochondrial reactive free oxygen species (MitSOX) in the glioblastoma cells. The aim was to elucidate the effects of CSP and MLT combination via the TRPM2 stimulation on the tumor cell viability, cell number, cell death (propidium iodide/Hoechst rate), apoptosis, and MitSOX levels in the DBTRG-05MG cells. In the DBTRG-05MG cells, we induced four groups as control, MLT (2.5 µg/ml for 24 h), CSP (25 µM for 24 h), and CSP + MLT. The CSP-induced intracellular Ca2+ influxes to the TRPM2 activation were increased in the cells from coming H2O2 and ADP-Ribose. The influxes were decreased in the cells by the incubations of TRPM2 antagonists (ACA and carvacrol). The incubation of CSP increased the parameters of intracellular Ca2+ responses, mitochondria function, cytosolic free Zn2+ accumulation, apoptosis (caspase -3, -8, and -9), and MitSOX generation in the tumor cells. After the treatment of MLT with/without CSP, the parameters were further increased in the cells. In conclusion, the treatment of MLT increased the anticancer, tumor cell death, apoptotic, and oxidant effects of CSP in the glioblastoma tumor cells via activating the TRPM2. As a result, TRPM2 stimulation by MLT may be utilized as a successful agent in the CSP treatment of glioblastoma tumors.


Subject(s)
Bee Venoms , Glioblastoma , TRPM Cation Channels , Humans , Cisplatin/pharmacology , Cisplatin/metabolism , Glioblastoma/drug therapy , Oxidative Stress , Melitten/pharmacology , TRPM Cation Channels/metabolism , Bee Venoms/pharmacology , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Reactive Oxygen Species/metabolism , Apoptosis , Oxidants/pharmacology , Calcium/metabolism
8.
Chem Biol Interact ; 369: 110261, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36403784

ABSTRACT

We investigated the effects of silver nanoparticle (AgNP) and cisplatin (CiSP) exposure via the activation of TRPM2 cation channels in glioblastoma (DBTRG-05MG) cell line. The cells were divided into four groups as control, AgNPs (100 µg/ml for 48 h), CiSP (25 µM for 24 h), and CiSP + AgNPs. We found that the cytotoxic, oxidant and apoptotic actions of CiSP were further stimulated through the activation of TRPM2 (via ADP-ribose and H2O2) in the cells by the treatment of AgNPs. The actions were decreased in the cells by the treatments of TRPM2 antagonists (ACA and 2APB). The apoptotic actions of AgNPs were induced by the stimulation of propidium iodide positive DBTRG-05MG rate, caspase -3, caspase -8, and caspase -9 activations, although their oxidant actions were acted by the increase of mitochondrial membrane depolarization, lipid peroxidation, mitochondrial oxygen free radicals (ROS), and cytosolic ROS, but the decrease of total antioxidant status, glutathione, and glutathione peroxidase. The accumulation of cytosolic free Ca2+ and Zn2+ into mitochondria via the activation of TRPM2 current density and activity accelerated oxidant and apoptotic actions of AgNPs in the cells. We found that the combination of AgNPs and CiSP was synergistic via the stimulation of TRPM2 for treatment of DBTRG-05MG cells. The combination of AgNPs and CiSP showed a favorable action via the stimulation of TRPM2 in the treatment of glioblastoma tumor cells.


Subject(s)
Glioblastoma , Metal Nanoparticles , TRPM Cation Channels , Humans , Cisplatin/pharmacology , Cisplatin/metabolism , Reactive Oxygen Species/metabolism , Oxidative Stress , TRPM Cation Channels/metabolism , Silver/pharmacology , Glioblastoma/drug therapy , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Apoptosis , Oxidants/pharmacology , Calcium/metabolism
9.
Biol Trace Elem Res ; 201(5): 2377-2395, 2023 May.
Article in English | MEDLINE | ID: mdl-36567422

ABSTRACT

Excessive levels of the mitochondrial reactive oxygen radical (mitSOX) and Ca2+ influx were found to cause neuropathic pain in patients with diabetes mellitus (DM). Naltriben (NLT) and mitSOX activate the transient receptor (TRP) melastatin 7 (TRPM7) channel, but antioxidants and carvacrol inhibit it. Selenium (Se) and curcumin (CRC) have been thoroughly studied for their modulator effects on streptozotocin (STZ)-induced neuropathic pain, apoptosis, and oxidative stress through the blockage of TRP channels in dorsal root ganglion (DRG) neurons. It has not yet been fully understood how Se and CRC protect against STZ-induced neuropathic pain by modulating TRPM7. Here, we assessed how Se and CRC affected the Ca2+ influx, mitSOX-mediated oxidative damage, and apoptosis in the DRGs of mice through modifying TRPM7 activity. Seven groups (control, Se, CRC, STZ, STZ + Se, STZ + CRC, and STZ + Se + CRC) were induced from the 56 male mice. We observed that the STZ-induced stimulation of TRPM7 increased mechanical neuropathic pain (von Frey), thermal neuropathic pain (hot plate), cytosolic Ca2+, TRPM7 current density, TRPM7 expression, lipid peroxidation, mitSOX, cytosolic ROS, apoptosis, caspase-3, caspase-8, and caspase-9 concentrations, whereas Se and CRC therapies diminished the alterations. The STZ-mediated decreases of DRG viability, brain glutathione, glutathione peroxidase, vitamin A, and vitamin E concentrations were also upregulated in the treatment groups by the therapies. These findings collectively imply that an imbalance of neuropathic pain, oxidative neurotoxicity, and apoptosis in the mice is caused by the STZ-mediated activation of TRPM7. However, the downregulation of TRPM7 activity caused by the injections of Se and CRC reduced the neurotoxicity and apoptosis.


Subject(s)
Curcumin , Diabetes Mellitus , Neuralgia , Selenium , TRPM Cation Channels , Mice , Male , Animals , Selenium/pharmacology , Curcumin/pharmacology , Curcumin/therapeutic use , TRPM Cation Channels/metabolism , Antioxidants/metabolism , Oxidative Stress , Streptozocin , Neuralgia/drug therapy , Neuralgia/metabolism
10.
Mol Biol Rep ; 50(3): 2207-2220, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36565417

ABSTRACT

BACKGROUND: Increased Ca2+ entry causes an increase in tumor cell proliferation, apoptosis, cytosolic reactive free oxygen species (cyROS), and mitochondrial ROS (miROS) in tumor cells. The cyROS and miROS stimulate the cation channels, including the TRPA1, TRPM2, and TRPV1. Sambucus ebulus L (SEB) (Dwarf Elder) induced both antioxidant and anticancer effects in the human hepatocarcinoma and human colon carcinoma cancer cell lines. We investigated the etiology of colorectal cancer and the impact of three channels, as well as the protective effects of SEB on apoptosis, cyROS, and miROS in the colon of mice with colitis-associated colon cancer (AOM/DSS). METHODS: A total 28 mice were equally divided into four groups as control, SEB (100 mg/kg/day for 14 days), AOM/DSS, and SEB + AOM/DSS. Azoxymethane/dextran sulfate sodium-induced colon cancer associated with colitis was induced in the AOM/DSS groups within 10 weeks. At the end of the experiments, the colon samples were removed from the mice. RESULTS: The protein bands of caspase - 3, TRPA1, TRPM2, and TRPV1 were increased by the treatments of AOM/DSS. The levels of apoptosis, cyROS, cleaved caspase - 3, and cleaved caspase - 9, as well as the depolarization of the mitochondrial membrane, all increased in the AOM/DSS group. Although they were reduced in the SEB and AOM/DSS + SEB groups by the treatments of SEB, TRPA1 (AP18), TRPM2 (ACA), and TRPV1 (capsazepine) antagonists, the apoptotic and oxidant values were further elevated in the AOM/DSS group by the treatments of TRPA1 (cinnamaldehyde), TRPM2 (H2O2), and TRPV1 (capsaicin) agonists. CONCLUSION: The activations of TRPA1, TRPM2, and TRPV1 channels induced the increase of apoptotic and oxidant actions in the colon cancer cells, although their inhibition via SEB treatment decreased the actions. Hence, TRPA1, TRPM2, and TRPV1 activations could be used as effective agents in the treatment of colon tumors.


Subject(s)
Colitis-Associated Neoplasms , TRPM Cation Channels , Transient Receptor Potential Channels , Mice , Humans , Animals , Transient Receptor Potential Channels/metabolism , TRPM Cation Channels/metabolism , Hydrogen Peroxide/metabolism , TRPV Cation Channels/metabolism , Oxidative Stress , Apoptosis , Reactive Oxygen Species/metabolism , Oxidants , Dextran Sulfate/pharmacology
11.
Cell Mol Neurobiol ; 43(3): 1335-1353, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35840808

ABSTRACT

Alzheimer's disease (AD) is characterized by the increase of hippocampal Ca2+ influx-induced apoptosis and mitochondrial oxidative stress (OS). The OS is a stimulator of TRPM2, although N-(p-amylcinnamoyl)anthranilic acid (ACA), 2-aminoethyl diphenylborinate (2/APB), and glutathione (GSH) are non-specific antagonists of TRPM2. In the present study, we investigated the protective roles of GSH and TRPM2 antagonist treatments on the amyloid ß42 peptide (Aß)-caused oxidative neurotoxicity and apoptosis in the hippocampus of mice with AD model. After the isolation of hippocampal neurons from the newborn mice, they were divided into five incubation groups as follows: control, ACA, Aß, Aß+ACA, and Aß+GSH. The levels of apoptosis, hippocampus death, cytosolic ROS, cytosolic Zn2+, mitochondrial ROS, caspase-3, caspase-9, lipid peroxidation, and cytosolic Ca2+ were increased in the primary hippocampus cultures by treatments of Aß, although their levels were decreased in the neurons by the treatments of GSH, PARP-1 inhibitors (PJ34 and DPQ), and TRPM2 blockers (ACA and 2/APB). The Aß-induced decreases of cell viability, cytosolic GSH, reduced GSH, and GSH peroxidase levels were also increased in the groups of Aß+ACA and Aß+GSH by the treatments of ACA and GSH. However, the Aß-caused changes were not observed in the hippocampus of TRPM2-knockout mice. In conclusion, the present data demonstrate that maintaining the activation of TRPM2 is not only important for the quenching OS and neurotoxicity in the hippocampal neurons of mice with experimental AD but also equally critical to the modulation of Aß-induced apoptosis. The possible positive effects of GSH and TRPM2 antagonist treatments on the amyloid-beta (Aß)-induced oxidative toxicity in the hippocampus of mice. The ADP-ribose (ADPR) is produced via the stimulation of PARP-1 in the nucleus of neurons. The NUT9 in the C terminus of TRPM2 channel acts as a key role for the activation of TRPM2. The antagonists of TRPM2 are glutathione (GSH), ACA, and 2/APB in the hippocampus. The Aß incubation-mediated TRPM2 stimulation increases the concentration of cytosolic-free Ca2+ and Zn2+ in the hippocampus. In turn, the increased concentration causes the increase of mitochondrial membrane potential (ΔΨm), which causes the excessive generations of mitochondria ROS and the decrease of cytosolic GSH and GSH peroxidase (GSH-Px). The ROS production and GSH depletion are two main causes in the neurobiology of Alzheimer's disease. However, the effect of Aß was not shown in the hippocampus of TRPM2-knockout mice. The Aß and TRPM2 stimulation-caused overload Ca2+ entry cause apoptosis and cell death via the activations of caspase-3 (Casp/3) and caspase-9 (Casp/9) in the hippocampus. The actions of Aß-induced oxidative toxicity were modulated in the primary hippocampus by the incubations of ACA, GSH, 2/APB, and PARP-1 inhibitors (PJ34 and DPQ). (↑) Increase. (↓) Decrease.


Subject(s)
Alzheimer Disease , TRPM Cation Channels , Rats , Mice , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Caspase 9/pharmacology , Reactive Oxygen Species/metabolism , TRPM Cation Channels/metabolism , Alzheimer Disease/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rats, Wistar , Oxidative Stress , Apoptosis , Glutathione/metabolism , Glutathione/pharmacology , Hippocampus/metabolism , Peroxidases/metabolism , Peroxidases/pharmacology , Mice, Knockout , Calcium/metabolism
13.
Behav Brain Res ; 434: 114033, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35914635

ABSTRACT

Chronic stress plays a key role in inducing various clinical disorders through mechanistic pathways, including oxidative stress and apoptosis. Transient receptor potential vanilloid 1 (TRPV1) channels, which are permeable to cations, mainly Ca2+, are susceptible to oxidative stress. Agomelatine (AGOM) is an antidepressant drug analogous to the antioxidant melatonin hormone, although its action has not been fully clarified yet. We aimed to investigate the protective role of AGOM on TRPV1-induced Ca2+ signaling and apoptosis in rats with chronic mild stress (CMS). The rats were divided into six main groups: control, dimethyl sulfoxide (DMSO), AGOM, CMS, CMS+DMSO, and CMS+AGOM. Five weeks of CMS were applied to rats in the CMS groups. The induction of CMS was confirmed with the sucrose preference test. The AGOM treatments were administered in the last three weeks of the experiment. The depression-like behavior, TRPV1-mediated cytosolic Ca2+ influx, lipid peroxidation, apoptosis, caspase - 3, and - 9 levels increased in the hippocampal neurons of CMS groups, although cell viability level was diminished by the CMS exposure. However, AGOM treatment downregulated stress-related behaviors, hippocampal oxidant and apoptotic markers by modulating the TRPV1 activity. In conclusion, TRPV1-mediated Ca2+ signaling and apoptosis may play a role in the etiopathogenesis of experimental depression. By regulating these changes with AGOM treatment, a positive contribution may be made to depression treatment.


Subject(s)
TRPM Cation Channels , Transient Receptor Potential Channels , Acetamides , Animals , Apoptosis , Calcium , Calcium Signaling , Depression , Dimethyl Sulfoxide , Hippocampus , Naphthalenes , Neurons , Oxidative Stress , Rats , TRPV Cation Channels
14.
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
15.
Graefes Arch Clin Exp Ophthalmol ; 260(8): 2567-2583, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35704089

ABSTRACT

PURPOSE: The concentration of plasma high glucose (HGu) in diabetes mellitus (DM) induces the retinal pigment epithelial cell (ARPE19) death via the increase of inflammation, cytosolic (cytROS), and mitochondrial (mitROS) free oxygen radical generations. Transient potential melastatin 2 (TRPM2) cation channel is stimulated by cytROS and mitROS. Hence, the cytROS and mitROS-mediated excessive Ca2+ influxes via the stimulation of TRPM2 channel cause to the induction of DM-mediated retina oxidative cytotoxicity. Because of the antioxidant role of carvacrol (CRV), it may modulate oxidative cytotoxicity via the attenuation of TRPM2 in the ARPE19. We aimed to investigate the modulator action of CRV treatment on the HGu-mediated TRPM2 stimulation, oxidative stress, and apoptosis in the ARPE19 cell model. MATERIAL AND METHODS: The ARPE19 cells were divided into four groups as normal glucose (NGu), NGu + Carv, HGu, and HGu + CRV. RESULTS: The levels of cell death (propidium iodide/Hoechst rate) and apoptosis markers (caspases 3, 8, and 9), cytokine generations (IL-1ß and TNF-α), ROS productions (cytROS, mitROS, and lipid peroxidation), TRPM2 currents, and intracellular free Ca2+ (Fluo/3) were increased in the HGu group after the stimulations of hydrogen peroxide and ADP-ribose, although their levels were diminished via upregulation of glutathione and glutathione peroxidase by the treatments of CRV and TRPM2 blockers. CONCLUSION: Current results confirmed that the HGu-induced overload Ca2+ influx and oxidative retinal toxicity in the ARPE19 cells were induced by the stimulation of TRPM2, although they were modulated via the inhibition of TRPM2 by CRV. CRV may be noted as a potential therapeutic antioxidant to the TRPM2 activation-mediated retinal oxidative injury.


Subject(s)
TRPM Cation Channels , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , Calcium , Cymenes , Epithelial Cells/metabolism , Ganglia, Spinal/metabolism , Glucose/toxicity , Humans , Inflammation/metabolism , Oxidative Stress/physiology , Retinal Pigments/metabolism , Signal Transduction , TRPM Cation Channels/metabolism
16.
Ocul Immunol Inflamm ; 30(6): 1315-1328, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35587813

ABSTRACT

PURPOSE: We investigated the possible protective effects of rituximab (RTX) on LPS-induced oxidant, inflammatory, and apoptotic adverse actions via the inhibition of TRPM2 channel in the adult retinal pigment epithelial-19 (ARPE-19) cells. METHODS: In the cultured ARPE-19 cells, we induced five main groups as control, RTX (10 µg/ml), LPS (1 µg/ml), LPS+RTX, and LPS+TRPM2 blockers (ACA or 2/APB). RESULTS: The levels of apoptosis, cell death, mitochondrial free reactive oxygen radicals (mitROS), cytosolic ROS, lipid peroxidation, caspase -3, caspase -8, caspase -9, ADP-ribose-induced TRPM2 current density, TNF-α, IL-1ß, cytosolic free Zn2+, and Ca2+ were increased by LPS, although their levels were diminished by the treatments of RTX and TRPM2 blockers. CONCLUSIONS: The LPS-induced mitROS, inflammatory cytokine, and apoptosis levels were modulated via TRPM2 inhibition in the human retinal epithelial cells by the RTX treatment. The RTX may be considered as a new therapeutic approach to LPS-induced human retinal epithelial cell injury.


Subject(s)
Lipopolysaccharides , TRPM Cation Channels , Humans , Lipopolysaccharides/toxicity , TRPM Cation Channels/metabolism , Rituximab/pharmacology , Apoptosis , Signal Transduction , Oxidative Stress , Inflammation/chemically induced , Inflammation/drug therapy , Retina
17.
Biol Trace Elem Res ; 200(5): 2283-2297, 2022 May.
Article in English | MEDLINE | ID: mdl-35384580

ABSTRACT

Diabetes mellitus induces optic nerve injury via the excessive generation of mitochondria reactive free oxygen radical (mitROS). TRPM2 channel is activated by mitROS, although it is inhibited by selenium (Se) and resveratrol (RSV). The activation of TRPM2 induces apoptosis and oxidative injury in the optic nerve. The inhibition of TRPM2 may decrease the optic nerve injury action of diabetes mellitus after the treatments of Se and RSV. Present study aimed to investigate the protective actions of Se and RSV on the excessive Ca2+ influx and mitROS generation-mediated optic nerve oxidative injury via the modulation of TRPM2. Fifty-six C57BL/6j male mice were divided into seven groups as control, Se, RSV, streptozotocin (STZ), STZ + Se, STZ + RSV, and STZ + Se + RSV. The STZ-mediated stimulation of TRPM2 increased the cytosolic Ca2+, lipid peroxidation, mitROS, cytosolic ROS, apoptosis, caspase-3, caspase-8, and caspase-9 concentrations in the mice, although their concentrations were decreased in the optic nerve by the treatments of Se and RSV. The STZ-induced decrease of optic nerve viability, glutathione, glutathione peroxidase, vitamin A, and vitamin E concentrations was also upregulated by the treatments of Se and RSV. The STZ-induced increase of TRPM2, PARP-1, caspase-3, and caspase-9 protein band expressions was diminished by the treatments of Se and RSV. In conclusion, STZ induced the optic nerve oxidative injury and apoptosis via the upregulation of TRPM2 stimulation, although the treatments of Se and RSV decreased the injury and apoptosis via the downregulation of TRPM2 activity.


Subject(s)
Diabetes Mellitus , Optic Nerve Injuries , Retinal Diseases , Selenium , TRPM Cation Channels , Animals , Apoptosis , Calcium/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Reactive Oxygen Species/metabolism , Resveratrol/pharmacology , Selenium/metabolism , Selenium/pharmacology , TRPM Cation Channels/metabolism
18.
Chem Biol Interact ; 359: 109914, 2022 May 25.
Article in English | MEDLINE | ID: mdl-35395232

ABSTRACT

Cisplatin (CiSP) induced-overload Ca2+ entry results in the increase of mitochondrial oxidative stress and apoptosis in the cancer cell. TRPM2 cation channel is gated by the cytosolic ADP-ribose (ADPR) and reactive oxygen species (ROS). The high content of polyunsaturated fatty acid (PUFA) in the brain is a main target of ROS. Eicosapentaenoic acid (EPA) induces oxidant action via the enhance of PUFA content in the glioblastoma (DBTRG) cells. We hypothesized that a combination of CiSP and EPA may offer a potential therapy in the DBTRG cell by exerting the antitumor, oxidant, and apoptotic actions and stimulating Ca2+ influx and TRPM2 activity. In the DBTRG cells, we induced four groups as control, EPA (30 µM for 24 h), CiSP (25 µM for 24 h), and CiSP + EPA. The CiSP-induced intracellular Ca2+ responses to the TRPM2 activation were increased in the DBTRG cells from coming H2O2 and ADPR. The responses were decreased in the cells by the inhibitions of TRPM2 (ACA and 2/APB) and PARP/1 (DPQ and PJ34). The incubation of EPA further increased the intracellular Ca2+ responses, mitochondria function, and the generation of ROS in the DBTRGs. After the treatment of EPA, lipid peroxidation, apoptosis, cell death, caspase -3, -8, and -9 levels were further increased in the DBTRG, although the levels of glutathione, glutathione peroxidase, cell numbers, and cell viability were further decreased in the cells. In summary, anticancer, apoptotic, and oxidant actions of CiSP were further increased via the activation of TRPM2 channel in the DBTRGs by the treatment of EPA. Hence, TRPM2 stimulation via EPA could be used as an effective agent in the treatment of glioblastoma tumors with CiSP.


Subject(s)
Brain Neoplasms , Glioblastoma , TRPM Cation Channels , Brain , Brain Neoplasms/drug therapy , Calcium/metabolism , Cell Count , Cisplatin/pharmacology , Eicosapentaenoic Acid/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Oxidative Stress , Reactive Oxygen Species/metabolism , TRPM Cation Channels/metabolism
19.
Mol Neurobiol ; 59(6): 3703-3720, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35366734

ABSTRACT

The hypoxia (HPX) acts the brain injury and apoptosis via the Ca2+ influx-induced excessive mitochondria free reactive oxygen species (mitROS) in neurons. The effective treatment of HPX is not possible yet. In addition to the antiviral and antiparkinsonian actions, amantadine (AMN) has been evaluated as a drug in treatments against brain injury. TRPM2 and TRPV4 channels are activated by mitROS. AMN attenuates NMDA receptor-induced Ca2+ influx, mitROS, inflammation, and apoptosis in the brain. However, the molecular pathways underlying AMN's neuroprotection against HPX remain elusive. We investigated the protective role of AMN via attenuation of TRPM2 and TRPV4 on oxidative neurotoxicity, mitochondrial membrane potential (ΔΨm), inflammation, and apoptosis in neuronal cells (SH-SY5Y). The SH-SY5Y and HEK293 cells were divided into six groups as follows: control, AMN (750 µM for 48 h), HPX (200 µM CoCl2 for 24 h), HPX + AMN, HPX + TRPM2 blockers (25 µM ACA or 100 µM 2APB for 30 min), and HPX + TRPV4 blocker (ruthenium red (RuR)-1 µM for 30 min). The HPX caused to upregulation of Ca2+ influx with an upregulation of ΔΨm and mitROS. The changes were not observed in the absence of TRPM2 and TRPV4 in the HEK293 cells. When HPX induction, TRPV4 agonist (GSK1016790A) and TRPM2 agonists (ADP-ribose and H2O2)-induced channel activity were diminished by the incubation of AMN and channel antagonists (RuR, ACA, and 2APB). The changes of mitROS, apoptotic markers (caspase-3 and -9), cell death rate, cell viability, cytokine (IL-1ß, IL-6, and TNF-α), ΔΨm, and Zn2+ concentrations were also restored by the incubation of AMN. In conclusion, the treatment of AMN attenuated HPX-mediated mitROS, apoptosis, and TRPM2/TRPV4-mediated overload Ca2+ influx and may provide an avenue for protecting the HPX-mediated neurodegenerative and cerebrovascular diseases associated with the upregulation of mitROS, Ca2+, and Zn2+ concentration.


Subject(s)
Brain Injuries , Neurotoxicity Syndromes , TRPM Cation Channels , Amantadine/pharmacology , Apoptosis , Calcium/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Hypoxia , Inflammation , Oxidative Stress , Reactive Oxygen Species/metabolism , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism
20.
Neurol Res ; 44(7): 636-644, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35019826

ABSTRACT

Parkinson's disease (PD) is an age-related chronic neurodegenerative disease. Although PD is known to be a result of damage to hippocampal neurons, its molecular mechanism has yet to be completely clarified. The neurodegeneration in hippocampal neurons has been suggested to include excessive production of reactive oxygen species (ROS). Mitochondrial dysfunction and disruption of intracellular Ca2+ homeostasis play the most important role in the increase in ROS production for the cells. Remarkably, it is stated in the literature that especially the change of Ca2+ homeostasis triggers neuronal degeneration. TRPM2 is a unique calcium-permeable nonselective cation channel, and densest in the numberless neuronal population. The current study aims to elucidate the effect of antioxidant resveratrol (Resv) on TRPM2-mediated oxidative stress (OS) induced by 1-methyl-4-phenylpyridinium (MPP) exposure in the primary mouse hippocampal neurons. The neurons were divided into four groups as Control, Resv , MPP, and MPP+ Resv. In the current results, the activation of TRPM2 was observed in primary hippocampal neurons with MPP incubation. TRPM2 channel expression levels in the MPP group increased in hippocampal neurons after MPP exposure. In addition, intracellular free Ca2+ concentration and TRPM2 channel currents were highest in MPP groups, although they were decreased by the Resv treatment. In addition, mitochondrial membrane depolarization, ROS, caspase-3, caspase-9, and apoptosis values induced by MPP decreased with resveratrol treatment. In conclusion, in our study, we observed that the dysregulation of OS-induced TRPM2 channel activation in hippocampal neurons exposed to MPP caused apoptotic cell death in neurons, while the use of resveratrol had a protective effect by reducing OS resources in the environment.


Subject(s)
Neurodegenerative Diseases , Neurons , Resveratrol , TRPM Cation Channels , 1-Methyl-4-phenylpyridinium , Animals , Animals, Newborn , Apoptosis , Hippocampus/cytology , Mice , Neurodegenerative Diseases/metabolism , Neurons/drug effects , Oxidative Stress , Reactive Oxygen Species/metabolism , Resveratrol/pharmacology , TRPM Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...