Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 255
Filter
1.
Front Immunol ; 15: 1294357, 2024.
Article in English | MEDLINE | ID: mdl-38318185

ABSTRACT

TRPM2 is a Ca2+ permeable, non-selective cation channel in the plasma membrane that is involved in the innate immune response regulating, for example, chemotaxis in neutrophils and cytokine secretion in monocytes and macrophages. The intracellular adenine nucleotides ADP-ribose (ADPR) and 2'-deoxy-ADPR (2dADPR) activate the channel, in combination with their co-agonist Ca2+. Interestingly, activation of human TRPM2 (hsTRPM2) by 2dADPR is much more effective than activation by ADPR. However, the underlying mechanism of the nucleotides' differential effect on the channel is not yet fully understood. In this study, we performed whole-cell patch clamp experiments with HEK293 cells heterologously expressing hsTRPM2. We show that 2dADPR has an approx. 4-fold higher Ca2+ sensitivity than ADPR (EC50 = 190 and 690 nM). This allows 2dADPR to activate the channel at lower and thus physiological intracellular Ca2+ concentrations. Kinetic analysis of our data reveals that activation by 2dADPR is faster than activation by ADPR. Mutation in a calmodulin binding N-terminal IQ-like motif in hsTRPM2 completely abrogated channel activation by both agonists. However, mutation of a single amino acid residue (W1355A) in the C-terminus of hsTRPM2, at a site of extensive inter-domain interaction, resulted in slower activation by 2dADPR and neutralized the difference in rate of activation between the two agonists. Taken together, we propose a mechanism by which 2dADPR induces higher hsTRPM2 currents than ADPR by means of faster channel activation. The finding that 2dADPR has a higher Ca2+ sensitivity than ADPR may indicate that 2dADPR rather than ADPR activates hsTRPM2 in physiological contexts such as the innate immune response.


Subject(s)
Adenosine Diphosphate Ribose , TRPM Cation Channels , Humans , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/pharmacology , Calcium Signaling , HEK293 Cells , Kinetics , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
2.
Br J Pharmacol ; 181(4): 564-579, 2024 02.
Article in English | MEDLINE | ID: mdl-36694432

ABSTRACT

BACKGROUND AND PURPOSE: Platelet function during inflammation is dependent on activation by endogenous nucleotides. Non-canonical signalling via the P2Y1 receptor is important for these non-thrombotic functions of platelets. However, apart from ADP, the role of other endogenous nucleotides acting as agonists at P2Y1 receptors is unknown. This study compared the effects of ADP, Ap3A, NAD+ , ADP-ribose, and Up4A on platelet functions contributing to inflammation or haemostasis. EXPERIMENTAL APPROACH: Platelets obtained from healthy human volunteers were incubated with ADP, Ap3A, NAD+ , ADP-ribose, or Up4A, with aggregation and fibrinogen binding measured (examples of function during haemostasis) or before exposure to fMLP to measure platelet chemotaxis (an inflammatory function). In silico molecular docking of these nucleotides to the binding pocket of P2Y1 receptors was then assessed. KEY RESULTS: Platelet aggregation and binding to fibrinogen induced by ADP was not mimicked by NAD+ , ADP-ribose, and Up4A. However, these endogenous nucleotides induced P2Y1 -dependent platelet chemotaxis, an effect that required RhoA and Rac-1 activity, but not canonical PLC activity. Analysis of molecular docking of the P2Y1 receptor revealed distinct differences of amino acid interactions and depth of fit within the binding pocket for Ap3A, NAD+ , ADP-ribose, or Up4A compared with ADP. CONCLUSION AND IMPLICATIONS: Platelet function (aggregation vs motility) can be differentially modulated by biased-agonist activation of P2Y1 receptors. This may be due to the character of the ligand-binding pocket interaction. This has implications for future therapeutic strategies aimed to suppress platelet activation during inflammation without affecting haemostasis as is the requirement of current ant-platelet drugs. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.


Subject(s)
Blood Platelets , NAD , Humans , Molecular Docking Simulation , NAD/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Diphosphate/metabolism , Platelet Aggregation , Inflammation/metabolism , Fibrinogen/metabolism , Fibrinogen/pharmacology , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/pharmacology , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y12/metabolism
3.
FASEB J ; 38(1): e23391, 2024 01.
Article in English | MEDLINE | ID: mdl-38145327

ABSTRACT

Adipocytes play a key role in energy storage and homeostasis. Although the role of transcription factors in adipocyte differentiation is known, the effect of endogenous metabolites of low molecular weight remains unclear. Here, we analyzed time-dependent changes in the levels of these metabolites throughout adipocyte differentiation, using metabolome analysis, and demonstrated that there is a positive correlation between cyclic adenosine diphosphate ribose (cADPR) and Pparγ mRNA expression used as a marker of differentiation. We also found that the treatment of C3H10T1/2 adipocytes with cADPR increased the mRNA expression of those marker genes and the accumulation of triglycerides. Furthermore, inhibition of ryanodine receptors (RyR), which are activated by cADPR, caused a significant reduction in mRNA expression levels of the marker genes and triglyceride accumulation in adipocytes. Our findings show that cADPR accelerates adipocytic differentiation via RyR pathway.


Subject(s)
Adipocytes , Cyclic ADP-Ribose , Mice , Animals , Cyclic ADP-Ribose/metabolism , Adipocytes/metabolism , Transcription Factors/metabolism , PPAR gamma/metabolism , Metabolome , RNA, Messenger/genetics , Cell Differentiation , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/pharmacology , Adipogenesis/genetics , 3T3-L1 Cells
4.
Article in Chinese | MEDLINE | ID: mdl-36229210

ABSTRACT

Objective: To investigate the effect of arsenic and its main metabolites on the apoptosis of human lung adenocarcinoma cell line A549 and the expression of pro-apoptotic genes Bad and Bik. Methods: In October 2020, A549 cells were recovered and cultured, and the cell viability was detected by the cell counting reagent CCK-8 to determine the concentration and time of sodium arsenite exposure to A549. The study was divided into NaAsO(2) exposure groups and metobol: le expoure groups: the metabolite comparison groups were subdivided into the control group, the monomethylarsinic acid exposure group (60 µmol/L) , and the dimethylarsinic acid exposure group (60 µmol/L) ; sodium arsenite dose groups were subdivided into 4 groups: control group (0) , 20, 40, 60 µmol/L sodium arsenite NaAsO(2). Hoechst 33342/propidium iodide double staining (Ho/PI) was used to observe cell apoptosis and real-time quantitative polymerase chain reaction (qRT-PCR) was used to detect the expression levels of Bad and Bik mRNA in cells after exposure. Western blotting was used to detect the protein expressions of Bad, P-Bad-S112, Bik, cleaved Bik and downstream proteins poly ADP-ribose polymerase PARP1 and cytochrome C (Cyt-C) , using spectrophotometry to detect the activity changes of caspase 3, 6, 8, 9. Results: Compared with the control group, the proportion of apoptotic cells in the 20, 40, and 60 µmol/L NaAsO(2) dose groups increased significantly (P<0.01) , and the expression levels of Bad, Bik mRNA, the protein expression levels of Bad, P-Bad-S112, Bik, cleaved Bik, PARP1, Cyt-C were increased (all P<0.05) , and the activities of Caspase 3, 6, 8, and 9 were significantly increased with significantly differences (P<0.05) . Compared with the control group, the expression level of Bad mRNA in the DMA exposure group (1.439±0.173) was increased with a significant difference (P=0.024) , but there was no significant difference in the expression level of Bik mRNA (P=0.788) . There was no significant differences in the expression levels of Bad and Bik mRNA in the poison groups (P=0.085, 0.063) . Compared with the control group, the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to MMA were 0.696±0.023, 0.707±0.014, 0.907±0.031, 1.032±0.016, and there was no significant difference between the two groups (P=0.469, 0.669, 0.859, 0.771) ; the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to DMA were 0.698±0.030, 0.705±0.022, 0.908±0.015, 1.029±0.010, and there was no difference between the two groups (P=0.479, 0.636, 0.803, 0.984) . Conclusion: Sodium arsenite induces the overexpression of Bad and Bik proteins, initiates the negative feedback regulation of phosphorylated Bad and the degradation of Bik, activates the downstream proteins PARP1, Cyt-C and Caspase pathways, and mediates the apoptosis of A549 cells.


Subject(s)
Arsenic , Poisons , A549 Cells , Adenosine Diphosphate Ribose/pharmacology , Apoptosis , Apoptosis Regulatory Proteins , Arsenites , Cacodylic Acid/pharmacology , Caspase 3 , Caspases/pharmacology , Cytochromes c/pharmacology , Humans , Mitochondrial Proteins/pharmacology , Propidium/pharmacology , RNA, Messenger , Sincalide/pharmacology , Sodium Compounds , bcl-Associated Death Protein/metabolism
5.
PLoS One ; 17(10): e0275748, 2022.
Article in English | MEDLINE | ID: mdl-36288391

ABSTRACT

Negative air ions (NAIs) being bioactive and negative charged molecules may confer antioxidant and anti-inflammatory activity. We assessed the effect of NAIs on two inflammatory diseases in animal models including lipopolysaccharide (LPS) induced acute lung injury (ALI) and wound healing in diabetic rats. We used intra-tracheal infusion of LPS to induce ALI and made a full-thickness cutaneous wound in streptozotocin-induced diabetic female Wistar rats. We evaluated NAIs effects on reactive oxygen species amount, leukocyte infiltration, wound healing rate, western blot, and immunohistochemistry in the lungs of ALI and skin sections of wounds. Our data found NAIs exposed saline displayed higher antioxidant activity vs. non-exposed saline. NAIs exposure did not significantly affect arterial blood pressure and respiratory frequency in control and LPS treated groups. LPS increased leukocyte infiltration, caspase 3/Poly-ADP-ribose-polymerase-mediated apoptosis formation and decreased Beclin-1/LC3-II-mediated autophagy in lungs. NAIs exposure conferred pulmonary protection by depressed leukocyte infiltration and caspase 3/Poly-ADP-ribose-polymerase mediated apoptosis and enhanced LC3-II-mediated autophagy in LPS induced ALI. NAIs treatment resulted in a significantly accelerated wound closure rate, decreased erythrocyte accumulation and leukocyte infiltration mediated oxidative stress and inflammation, and upregulated expression of skin collagen, vascular endothelial growth factor receptor-2 (VEGFR-2) and factor transforming growth factor-beta 1 (TGF-ß1) vs non-treated group. Based on these results, it is suggested that NAIs conferred a protection through the upregulating LC3-II-dependent autophagy mechanism and downregulating leukocyte infiltration mediated inflammation and caspase 3/Poly-ADP-ribose-polymerase signaling in the LPS-treated ALI and promoted diabetic wound healing through the enhancing skin collagen synthesis, VEGFR-2 and TGF-ß1 pathways.


Subject(s)
Acute Lung Injury , Diabetes Mellitus, Experimental , Rats , Female , Animals , Lipopolysaccharides/pharmacology , Vascular Endothelial Growth Factor Receptor-2 , Antioxidants/pharmacology , Caspase 3 , Transforming Growth Factor beta1/pharmacology , Reactive Oxygen Species/pharmacology , Beclin-1 , Streptozocin/pharmacology , Diabetes Mellitus, Experimental/complications , Vascular Endothelial Growth Factor A/pharmacology , Rats, Wistar , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Wound Healing , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology , Ions , Transforming Growth Factors , Adenosine Diphosphate Ribose/pharmacology
6.
J Inorg Biochem ; 236: 111963, 2022 11.
Article in English | MEDLINE | ID: mdl-35988387

ABSTRACT

In this article, ligand IPP (IPP = 4-(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)-N,N-diphenylaniline) and its three Ru(II) complexes: [Ru(bpy)2(IPP)](ClO4)2 (1) (bpy = 2,2'-bipyridine), [Ru(dmbpy)2(IPP)](ClO4)2 (2) (dmbpy = 4,4'-dimethyl-2,2'-bipyridine), and [Ru(phen)2(IPP)](ClO4)2 (3) (phen = 1,10-phenanthroline) were synthesized and characterized. The anticancer activity in vitro of the complexes was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. The scratching and colony-forming experiments confirmed the complexes 1, 2, 3 interfered with the proliferation and migration ability of cells. The accumulation of the complexes in cells was researched and we found that these complexes directly accumulated in mitochondria, then the complexes cause a decline of the mitochondrial membrane potential and induce an increase of intracellular reactive oxygen species (ROS) levels. The growth of B16 cells were inhibited by 1, 2 and 3 at G0/G1 phase. Apoptosis was induced through mitochondrial pathway and the expression of apoptosis-related factors was regulated. In addition, the complexes promoted the transition of poly(ADP-ribose)polymerase (PARP) into the cleaved form (Cleaved PARP), downregulated the anti-apoptotic proteins, and upregulated the pro-apoptotic proteins. Consequently, complexes 1, 2 and 3 exerted their anticancer activity by regulating B-cell lymphoma-2 (Bcl-2) family proteins. Complex 2 showed excellent antitumor effects with a high inhibitory rate of 65.95% in vivo. Taken together, the complexes cause apoptosis in B16 cells through a ROS-mediated mitochondrial dysfunction pathway.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Ruthenium , 2,2'-Dipyridyl/pharmacology , Adenosine Diphosphate Ribose/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins , Cell Line, Tumor , Coordination Complexes/pharmacology , Ligands , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Ruthenium/pharmacology
7.
Pharmacology ; 107(9-10): 472-479, 2022.
Article in English | MEDLINE | ID: mdl-35584608

ABSTRACT

INTRODUCTION: The heat and redox-sensitive ion channel TRPM2 was reported to be a causative mechanism for depression in a mouse model and to be upregulated in the hippocampus in patients suffering from depressive disorders. TRPM2 may thus be a novel target for antidepressants, but so far, selective TRPM2-inhibitors have not yet been developed. In this in vitro study, we examined the inhibitory effects of several established antidepressants on heat-evoked inward currents of TRPM2. METHODS: Human (h) TRPM2 expressed in HEK293 cells was examined by means of whole-cell patch clamp recordings. Effects of duloxetine, amitriptyline, sertraline, fluoxetine, paroxetine, citalopram, escitalopram, ketamine, pregabalin, lidocaine, and QX-314 were explored on heat-evoked currents in cells pretreated with ADP-ribose (ADPR). RESULTS: While inward currents induced by 1 mM ADPR in the pipette solution displayed a strong rundown hampering pharmacological experiments, heat-evoked currents in cells loaded with 200 µM APDR remained stable upon repetitive activation. Among all substances examined, only inhibition induced by duloxetine displayed a clear concentration-dependency. Thirty micromolar duloxetine was required for 50% inhibition, the same degree of inhibition was also induced by 30 µM amitriptyline, fluoxetine, and paroxetine. While citalopram, escitalopram, ketamine, and pregabalin failed to robustly modify TRPM2, sertraline and low concentrations of lidocaine even potentiated heat-evoked currents. CONCLUSION: Our data indicate that some, but not all established antidepressants inhibit hTRPM2 when it is activated by heat and ADPR in vitro, e.g., presumably relevant endogenous agonists. However, none of the examined substances exhibited a potent inhibition which is likely to translate into a clinically relevant effect at effective plasma concentrations. Whether or not TRPM2 may be a relevant target for antidepressants cannot be conclusively assessed by a single in vitro study, thus further studies are required along these lines. Nevertheless, future studies may get simplified by the novel approach we developed for in vitro pharmacological analysis of TRPM2.


Subject(s)
Ketamine , TRPM Cation Channels , Adenosine Diphosphate Ribose/pharmacology , Amitriptyline/pharmacology , Animals , Antidepressive Agents/pharmacology , Citalopram/pharmacology , Duloxetine Hydrochloride/pharmacology , Fluoxetine/pharmacology , HEK293 Cells , Hot Temperature , Humans , Lidocaine/pharmacology , Mice , Paroxetine , Pregabalin , Sertraline
8.
Environ Toxicol ; 37(8): 1891-1901, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35396826

ABSTRACT

Methylmercury (MeHg) is an environmental neurotoxic substance, which can easily cross the blood-brain barrier, causing irreversible damage to the human central nervous system. Reactive oxygen species (ROS) are involved in various ways of intracellular physiological or pathological processes including neuronal apoptosis. This study attempted to explore the role of ROS-mediated poly ADP-ribose polymerase (PARP)/apoptosis-inducing factor (AIF) apoptosis signaling pathway in the process of MeHg-induced cell death of human neuroblastoma cells (SH-SY5Y). Here, we found that SH-SY5Y cells underwent apoptosis in response to MeHg, which was accompanied by the increased levels of ROS and calcium ion, and the activation of caspase cascades and PARP. Inhibiting the production of ROS can reduce the apoptosis rate to a certain extent. PARP/AIF apoptotic pathway is independent of caspase dependent signaling pathway and regulates it. In conclusion, these results suggest that ROS mediated activation of caspase pathway and PARP/AIF signaling pathway are involved in MeHg induced apoptosis, and these two pathways interact with each other.


Subject(s)
Methylmercury Compounds , Neuroblastoma , Adenosine Diphosphate Ribose/pharmacology , Apoptosis , Apoptosis Inducing Factor/metabolism , Apoptosis Inducing Factor/pharmacology , Caspases/metabolism , Humans , Methylmercury Compounds/toxicity , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism
9.
Acta Pharmacol Sin ; 43(4): 759-770, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34108651

ABSTRACT

Ion channels are ubiquitously expressed in almost all living cells, and are the third-largest category of drug targets, following enzymes and receptors. The transient receptor potential melastatin (TRPM) subfamily of ion channels are important to cell function and survival. Studies have shown upregulation of the TRPM family of ion channels in various brain tumours. Gliomas are the most prevalent form of primary malignant brain tumours with no effective treatment; thus, drug development is eagerly needed. TRPM2 is an essential ion channel for cell function and has important roles in oxidative stress and inflammation. In response to oxidative stress, ADP-ribose (ADPR) is produced, and in turn activates TRPM2 by binding to the NUDT9-H domain on the C-terminal. TRPM2 has been implicated in various cancers and is significantly upregulated in brain tumours. This article reviews the current understanding of TRPM2 in the context of brain tumours and overviews the effects of potential drug therapies targeting TRPM2 including hydrogen peroxide (H2O2), curcumin, docetaxel and selenium, paclitaxel and resveratrol, and botulinum toxin. It is long withstanding knowledge that gliomas are difficult to treat effectively, therefore investigating TRPM2 as a potential therapeutic target for brain tumours may be of considerable interest in the fields of ion channels and pharmacology.


Subject(s)
Brain Neoplasms , TRPM Cation Channels , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/pharmacology , Brain Neoplasms/drug therapy , Calcium/metabolism , Humans , Hydrogen Peroxide/pharmacology , Oxidative Stress , TRPM Cation Channels/physiology
10.
J Natl Cancer Inst ; 113(12): 1770-1778, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34048575

ABSTRACT

BACKGROUND: Loss of ovarian function is a recognized adverse effect of chemotherapy for breast cancer and of great importance to patients. Little is known about the ovarian toxicity of newer cancer treatments. This study examined whether breast cancer clinical trials include assessment of the impact of trial interventions on ovarian function. METHODS: Eligible trials were phase III (neo)adjuvant trials of pharmacologic treatments for breast cancer, recruiting between June 2008 and October 2019, which included premenopausal women. MEDLINE, EMBASE, Clinicaltrials.gov, and EudraCT were searched. Data were extracted from trial publications, protocols, databases, and a survey sent to all trial chairs. Tests of statistical significance were 2-sided. RESULTS: Of 2354 records identified, 141 trials were eligible. Investigational treatments included chemotherapy (36.9%), HER2 targeted (24.8%), endocrine (12.8%), immunotherapy (7.8%), cyclin-dependent kinase 4/6 inhibitors (5.0%), and poly-ADP-ribose polymerase inhibitors (2.8%). Ovarian function was a prespecified endpoint in 13 (9.2%) trials. Forty-five (31.9%) trials collected ovarian function data, but only 33 (23.4%) collected posttrial-intervention data. Common postintervention data collected included menstruation (15.6%), pregnancy (13.5%), estradiol (9.9%), and follicle-stimulating hormone levels (8.5%). Only 4 (2.8%) trials collected postintervention anti-müllerian hormone levels, and 3 (2.1%) trials collected antral follicle count. Of 22 trials investigating immunotherapy, cyclin-dependent kinase 4/6 inhibitors, or poly-ADP-ribose polymerase inhibitors, none specified ovarian function as an endpoint, but 4 (18.2%) collected postintervention ovarian function data. CONCLUSIONS: The impact of pharmacologic interventions on ovarian function is infrequently assessed in phase III breast cancer (neo)adjuvant trials that include premenopausal women. Trialists should consider inclusion of ovarian function endpoints when designing clinical trials, given its importance for informed decision making.


Subject(s)
Breast Neoplasms , Female , Humans , Pregnancy , Adenosine Diphosphate Ribose/pharmacology , Adenosine Diphosphate Ribose/therapeutic use , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4 , Ovary , Premenopause , Clinical Trials, Phase III as Topic
11.
Eur J Pharmacol ; 882: 173163, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32485244

ABSTRACT

Oxidative stress-induced Ca2+ permeable transient receptor potential melastatin 2 (TRPM2) channels are expressed at high levels in the brain, appear to link neuronal excitability to cellular metabolism, and are involved in the pathogenesis of neurodegenerative disorders. We aimed to study the electrophysiological properties of TRPM2 channels in stellate cells of the mouse ventral cochlear nucleus (VCN) using molecular, immunohistochemical and electrophysiological approaches. In the present study, the real time PCR analysis revealed the presence of the TRPM2 mRNA in the mouse VCN tissue. Cell bodies of stellate cells were moderately labeled with TRPM2 antibodies using immunohistochemical staining. Stellate cells were sensitive to intracellular ADP-ribose (ADPR), a TRPM2 agonist. Upon the application of ADPR, the resting membrane potential of the stellate cells was significantly depolarized, shifting from -61.2 ± 0.9 mV to -57.0 ± 0.8 mV (P < 0.001; n = 21), and the firing rate significantly increased (P < 0.001, n = 6). When the pipette solution contained ADPR (300 µM) and the TRPM2 antagonists flufenamic acid (FFA) (100 µM), N-(p-amylcinnamoyl) anthranilic acid (ACA) (50 µM) and 8-bromo-cADP-Ribose (8-Br-cADPR) (50 µM), the membrane potential shifted in a hyperpolarizing direction. ADPR did not significantly change the resting membrane potential and action potential firing rate of stellate cells from TRPM2-/- mice. In conclusion, the results obtained using these molecular, immunohistochemical and electrophysiological approaches reveal the expression of functional TRPM2 channels in stellate neurons of the mouse VCN. TRPM2 might exert a significant modulatory effect on setting the level of resting excitability.


Subject(s)
Cochlear Nucleus/physiology , Neurons/physiology , TRPM Cation Channels/physiology , Adenosine Diphosphate Ribose/pharmacology , Animals , Cochlear Nucleus/metabolism , Female , Male , Mice, Inbred BALB C , Mice, Knockout , Neurons/metabolism , TRPM Cation Channels/agonists , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
12.
Sci Rep ; 9(1): 4100, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858386

ABSTRACT

Activation of TRPM8 channel through oxidative stress may induce Ca2+ and pro-apoptotic signals in prostate cancer and kidney cells. The aim of this study was to evaluate activation of TRPM8 can increase apoptosis and oxidative stress in the prostate cancer (Du145M8), TRPM8 knock out (Du 145M8KO), transfected (HEK293TM8) and non-transfected human kidney (HEK293) cells. Intracellular Ca2+ responses to TRPM8 activation were increased in the Du145M8 and HEK293TM8 cells from coming cumene hydrogen peroxide (CHPx), menthol, ADP-Ribose (ADPR), but not in the HEK293 and Du 145M8KO cells. The intracellular Ca2+ responses to both ADPR and CHPx were totally inhibited by the thiol cycle antioxidant glutathione, and TRPM8 blockers (N-(p-amylcinnamoyl)anthranilic acid and capsazepine). Apoptosis, Annexin V, mitochondrial membrane depolarization, intracellular ROS, caspase 3 and 9 values were increased through TRPM8 activation in the Du 145M8 but not in the Du 145M8KO and non-transfected HEK293 cells by CHPx and hydrogen peroxide. In conclusion, apoptotic and oxidant effects on the cells were increased activation of TRPM8 by oxidative stress and ADPR. Activation of TRPM8 through oxidative stress and ADPR in the cells could be used as an effective strategy in the treatment of prostate cancer cells.


Subject(s)
Adenosine Diphosphate Ribose/pharmacology , Kidney/metabolism , Oxidative Stress/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , TRPM Cation Channels/metabolism , Annexin A5/metabolism , Apoptosis/drug effects , Calcium/metabolism , Cinnamates/pharmacology , Fluorescence , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , Ion Channel Gating/drug effects , Kidney/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , ortho-Aminobenzoates/pharmacology
13.
World Neurosurg ; 127: e346-e352, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30904799

ABSTRACT

BACKGROUND: Neuronal apoptosis plays a pivotal role in spinal cord injury (SCI)-induced secondary cellular events. Caspase-dependent and -independent pathways are involved in neuronal apoptosis. Caspase-3 is the final effector of caspase-dependent apoptosis, whereas poly-ADP-ribose polymerase-1 (PARP-1) and apoptosis-inducing factor (AIF) are key executors of caspase-independent apoptosis. However, it remains unclear whether simultaneous inhibition of the 2 apoptosis pathways will be more beneficial for neuronal survival. Therefore, this study investigated the ability of coadministration of the PARP-1 inhibitor 3-aminobenzamide (3-AB) and caspase-3 inhibitor z-DEVD-fmk to attenuate apoptosis in a rat SCI model. METHODS: The rats were subjected to moderate contusive SCI. Locomotor function was measured using the Basso, Beattie, and Bresnahan rating scales; neuronal apoptosis was detected using transferase-mediated deoxyuridine triphosphate-biotin nick end labeling; and immunohistochemistry and Western blotting were used to measure protein expression. RESULTS: We found the locomotor function of rats was weakened within 7 days post-SCI. At day 7 post-SCI, neuronal apoptosis dramatically increased and the expression of PARP-1, AIF, and cleaved caspase-3 was significantly upregulated. Further, Bcl-2 expression was significantly downregulated. The highest locomotor function recovery was recorded after the combined administration of 3-AB and z-DEVD-fmk for 7 days post-SCI when compared with 3-AB or z-DEVD-fmk administered alone. In addition, this combination therapy significantly reduced neuronal apoptosis by preventing upregulation of PARP-1 and AIF, inhibiting caspase-3 activation, and elevating Bcl-2 expression. CONCLUSIONS: These results suggest that combination therapy is beneficial for neuronal function recovery in rats with SCI. The underlying mechanism may be associated with cosuppression of caspase-dependent and caspase-independent apoptosis pathways.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors/pharmacology , Neurons/drug effects , Oligopeptides/pharmacology , Spinal Cord Injuries/drug therapy , Adenosine Diphosphate Ribose/pharmacology , Animals , Caspase 3/metabolism , Male , Neurons/metabolism , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
14.
Nature ; 562(7725): 145-149, 2018 10.
Article in English | MEDLINE | ID: mdl-30250252

ABSTRACT

Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable, non-selective cation channel that has an essential role in diverse physiological processes such as core body temperature regulation, immune response and apoptosis1-4. TRPM2 is polymodal and can be activated by a wide range of stimuli1-7, including temperature, oxidative stress and NAD+-related metabolites such as ADP-ribose (ADPR). Its activation results in both Ca2+ entry across the plasma membrane and Ca2+ release from lysosomes8, and has been linked to diseases such as ischaemia-reperfusion injury, bipolar disorder and Alzheimer's disease9-11. Here we report the cryo-electron microscopy structures of the zebrafish TRPM2 in the apo resting (closed) state and in the ADPR/Ca2+-bound active (open) state, in which the characteristic NUDT9-H domains hang underneath the MHR1/2 domain. We identify an ADPR-binding site located in the bi-lobed structure of the MHR1/2 domain. Our results provide an insight into the mechanism of activation of the TRPM channel family and define a framework for the development of therapeutic agents to treat neurodegenerative diseases and temperature-related pathological conditions.


Subject(s)
Adenosine Diphosphate Ribose/pharmacology , Calcium/pharmacology , TRPM Cation Channels/metabolism , TRPM Cation Channels/ultrastructure , Zebrafish Proteins/metabolism , Zebrafish Proteins/ultrastructure , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Animals , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Binding Sites , Calcium/chemistry , Calcium/metabolism , Cryoelectron Microscopy , Edetic Acid/chemistry , Humans , Ion Channel Gating/drug effects , Ligands , Models, Molecular , Neurodegenerative Diseases/drug therapy , Protein Domains , Pyrophosphatases/chemistry , Signal Transduction/drug effects , TRPM Cation Channels/chemistry , Zebrafish , Zebrafish Proteins/chemistry
15.
Nat Chem Biol ; 13(9): 1036-1044, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28671679

ABSTRACT

Transient receptor potential melastatin 2 (TRPM2) is a ligand-gated Ca2+-permeable nonselective cation channel. Whereas physiological stimuli, such as chemotactic agents, evoke controlled Ca2+ signals via TRPM2, pathophysiological stimuli such as reactive oxygen species and genotoxic stress result in prolonged TRPM2-mediated Ca2+ entry and, consequently, apoptosis. To date, adenosine 5'-diphosphoribose (ADPR) has been assumed to be the main agonist for TRPM2. Here we show that 2'-deoxy-ADPR was a significantly better TRPM2 agonist, inducing 10.4-fold higher whole-cell currents at saturation. Mechanistically, this increased activity was caused by a decreased rate of inactivation and higher average open probability. Using high-performance liquid chromatography (HPLC) and mass spectrometry, we detected endogenous 2'-deoxy-ADPR in Jurkat T lymphocytes. Consistently, cytosolic nicotinamide mononucleotide adenylyltransferase 2 (NMNAT-2) and nicotinamide adenine dinucleotide (NAD)-glycohydrolase CD38 sequentially catalyzed the synthesis of 2'-deoxy-ADPR from nicotinamide mononucleotide (NMN) and 2'-deoxy-ATP in vitro. Thus, 2'-deoxy-ADPR is an endogenous TRPM2 superagonist that may act as a cell signaling molecule.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Clusterin/agonists , ADP-ribosyl Cyclase 1/chemistry , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/pharmacology , Chromatography, High Pressure Liquid , Humans , Hydrogen Peroxide/chemistry , Jurkat Cells , Molecular Structure , Signal Transduction/drug effects
16.
J Gen Physiol ; 149(2): 219-235, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28108595

ABSTRACT

Activation of the transient receptor potential melastatin 2 (TRPM2) channel occurs during the response to oxidative stress under physiological conditions as well as in pathological processes such as ischemia and diabetes. Accumulating evidence indicates that adenosine diphosphate ribose (ADPR) is the most important endogenous ligand of TRPM2. However, although it is known that ADPR binds to the NUDT9 homology (NUDT9-H) domain in the intracellular C-terminal region, the molecular mechanism underlying ADPR binding and activation of TRPM2 remains unknown. In this study, we generate a structural model of the NUDT9-H domain and identify the binding pocket for ADPR using induced docking and molecular dynamics simulation. We find a subset of 11 residues-H1346, T1347, T1349, L1379, G1389, S1391, E1409, D1431, R1433, L1484, and H1488-that are most likely to directly interact with ADPR. Results from mutagenesis and electrophysiology approaches support the predicted binding mechanism, indicating that ADPR binds tightly to the NUDT9-H domain, and suggest that the most significant interactions are the van der Waals forces with S1391 and L1484, polar solvation interaction with E1409, and electronic interactions (including π-π interactions) with H1346, T1347, Y1349, D1431, and H1488. These findings not only clarify the roles of a range of newly identified residues involved in ADPR binding in the TRPM2 channel, but also reveal the binding pocket for ADPR in the NUDT9-H domain, which should facilitate structure-based drug design for the TRPM2 channel.


Subject(s)
Adenosine Diphosphate Ribose/pharmacology , Molecular Docking Simulation , TRPM Cation Channels/chemistry , Adenosine Diphosphate Ribose/chemistry , Amino Acid Substitution , Binding Sites , HEK293 Cells , Humans , Protein Binding , Pyrophosphatases/chemistry , Sequence Homology , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
17.
Sci Rep ; 6: 38471, 2016 12 23.
Article in English | MEDLINE | ID: mdl-28008929

ABSTRACT

TRPM2, one member of the transient receptor potential (TRP) protein super-family, is a Ca2+-permeable channel that is activated by oxidative stress and confers susceptibility to cell death. In the human tongue specimens of carcinoma and the tongue carcinoma SCC cell lines, we observed the enhanced expression of TRPM2. By means of the whole-cell electrophysiological recording, the ADPR-induced currents mediated by TRPM2 were recorded in cultured SCC9 cells. Moreover, after H2O2 treatment for 24 hours, the apoptotic number of SCC9 cells was significantly increased. However, the selectively knocked-down TRPM2 with the small interfering RNA technique inhibited the survival and migration of the SCC9 cancer cells, which was independent of the p53-p21 pathway, since the expression of p21 was enhanced after TRPM2 knockdown. Furthermore, the sub-cellular localization of TRPM2 was remarkably different between cancerous and non-cancerous cells. A significant amount of the TRPM2 proteins were located in the nuclei in cancer cells. All these data suggest that TRPM2 is essential for the survival and migration of SCC cancer cells and may be a potential target for the selective treatment of tongue cancer.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Clusterin/metabolism , Mouth Neoplasms/metabolism , Adenosine Diphosphate Ribose/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Hydrogen Peroxide/pharmacology , Ion Channel Gating/drug effects , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Oxidative Stress/drug effects , Subcellular Fractions/metabolism , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Up-Regulation/drug effects , Up-Regulation/genetics
18.
PLoS One ; 11(6): e0158060, 2016.
Article in English | MEDLINE | ID: mdl-27333281

ABSTRACT

The human redox-sensitive Transient receptor potential melastatin type 2 (hTRPM2) channel contains the C-terminal Nudix hydrolase domain NUDT9H which most likely binds ADP-ribose. During oxidative stress, the intracellular release of ADP-ribose triggers the activation of hTRPM2. The TRPM2 orthologue from Nematostella vectensis (nv) is also stimulated by ADP-ribose but not by the oxidant hydrogen peroxide. For further clarification of the structure-function relationships of these two distantly related channel orthologues, we performed whole-cell as well as single channel patch-clamp recordings, Ca2+-imaging and Western blot analysis after heterologous expression of wild-type and mutated channels in HEK-293 cells. We demonstrate that the removal of the entire NUDT9H domain does not disturb the response of nvTRPM2 to ADP-ribose. The deletion, however, created channels that were activated by hydrogen peroxide, as did mutations within the NUDT9H domain of nvTRPM2 that presumably suppress its enzymatic function. The same findings were obtained with the nvTRPM2 channel when the NUDT9H domain was replaced by the corresponding sequences of the original hNUDT9 enzyme. Whenever the enzyme domain was mutated to presumably inactive variants, channel activation by hydrogen peroxide could be achieved. Moreover, we found strong evidences for ADPRase activity of the isolated NUDT9H domain of nvTRPM2 in co-expression experiments with the C-terminally truncated nvTRPM2 channel. Thus, there is a clear correlation between the loss of enzymatic activity and the capability of nvTRPM2 to respond to oxidative stress. In striking contrast, the channel function of the hTRPM2 orthologue, in particular its sensitivity to ADP-ribose, was abrogated by already small changes of the NUDT9H domain. These findings establish nvTRPM2 as a channel gated by ADP-ribose through a novel mechanism. We conclude that the endogenous NUDT9H domain does not directly affect ADP-ribose-dependent gating of the nvTRPM2 channel; instead it exerts an independent catalytic function which possibly controls the intracellular availability of ADP-ribose.


Subject(s)
Adenosine Diphosphate Ribose/pharmacology , Ion Channel Gating/drug effects , Sea Anemones/metabolism , TRPM Cation Channels/chemistry , TRPM Cation Channels/metabolism , Amino Acid Sequence , Animals , Biocatalysis/drug effects , Blotting, Western , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , Models, Biological , Mutant Proteins/metabolism , Mutation/genetics , Patch-Clamp Techniques , Protein Domains , Sequence Deletion , TRPM Cation Channels/genetics
19.
Cell Physiol Biochem ; 37(5): 2043-59, 2015.
Article in English | MEDLINE | ID: mdl-26584302

ABSTRACT

BACKGROUND/AIMS: Adenosine diphosphate ribose (ADPR), a product of ß-NAD+ metabolism generated by the multifunctional enzyme CD38, is recognized as a novel signaling molecule. The catalytic site of CD38 orients extracellularly or intracellularly, capable of generating ADPR outside and inside the cells. CD38-dependent pathways have been characterized in pulmonary artery smooth muscle cells (PASMCs); however the physiological function of extracellular ADPR is unclear. METHODS: Ca2+ mobilizing and proliferative effects of extracellular ADPR were characterized and compared with the ATP-induced responses in rat PASMCs; and the expression of purinergic receptor (P2X and P2Y) subtypes were examined in pulmonary arteries. RESULTS: ADPR elicited concentration-dependent increase in [Ca2+]i with a fast transient and a sustained phase in PASMCs. The sustained phase was abolished by Ca2+ removal and inhibited by the non-selective cation channel blocker SKF-96365, but was unaffected by TRPM2 antagonists or nifedipine. The purinergic receptor (P2X) antagonist pyridoxal-phosphate-6-azophenyl-2', 4'-disulfonate inhibited partially the transient and the sustained Ca2+ response, while the P2(XY) inhibitor suramin and the phospholipase C inhibitor U73122 abolished the sustained Ca2+ influx. The P2Y1 antagonist MRS2179 had no effect on the response. By contrast, ATP and ADP activated Ca2+ response exhibited a high and a low affinity component, and the pharmacological profile of ATP-induced Ca2+ response was distinctive from that of ADPR. BrdU incorporation assay showed that ADPR caused significant inhibition whereas ATP caused slight stimulation of PASMC proliferation. RT-PCR analysis found that almost all P2X and P2Y subtypes are expressed in PAs. CONCLUSION: ADPR and ATP activate Ca2+ responses through different combinations of multiple purinergic receptor subtypes; and extracellular ADPR may exert an autocrine/paracrine action via purinergic receptors on PASMCs.


Subject(s)
Adenosine Diphosphate Ribose/pharmacology , Calcium Signaling/drug effects , Calcium/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Estrenes/pharmacology , Imidazoles/pharmacology , Ions/chemistry , Ions/metabolism , Male , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nifedipine/pharmacology , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Pyrrolidinones/pharmacology , Rats , Rats, Wistar , Receptors, Purinergic P2X/chemistry , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2Y1/chemistry , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Signal Transduction/drug effects , Suramin/pharmacology , TRPM Cation Channels/metabolism , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...