Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.129
Filter
1.
Molecules ; 29(17)2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39274863

ABSTRACT

A Fe2+-EGTA(ethylene glycol-bis (ß-aminoethyl ether)-N,N,N',N'-tetraacetic acid)-H2O2 system emits photons, and quenching this chemiluminescence can be used for determination of anti-hydroxyl radical (•OH) activity of various compounds. The generation of •OH and light emission due to oxidative damage to EGTA may depend on the buffer and pH of the reaction milieu. In this study, we evaluated the effect of pH from 6.0 to 7.4 (that may occur in human cells) stabilized with 10 mM phosphate buffer (main intracellular buffer) on a chemiluminescence signal and the ratio of this signal to noise (light emission from medium alone). The highest signal (4698 ± 583 RLU) and signal-to-noise ratio (9.7 ± 1.5) were noted for pH 6.6. Lower and higher pH caused suppression of these variables to 2696 ± 292 RLU, 4.0 ± 0.8 at pH 6.2 and to 3946 ± 558 RLU, 5.0 ± 1.5 at pH 7.4, respectively. The following processes may explain these observations: enhancement and inhibition of •OH production in lower and higher pH; formation of insoluble Fe(OH)3 at neutral and alkaline environments; augmentation of •OH production by phosphates at weakly acidic and neutral environments; and decreased regeneration of Fe2+-EGTA in an acidic environment. Fe2+-EGTA-H2O2 system in 10 mM phosphate buffer pH 6.6 seems optimal for the determination of anti-•OH activity.


Subject(s)
Egtazic Acid , Hydrogen Peroxide , Hydrogen Peroxide/chemistry , Hydrogen-Ion Concentration , Humans , Egtazic Acid/chemistry , Egtazic Acid/analogs & derivatives , Hydroxyl Radical/chemistry , Iron/chemistry , Luminescence , Luminescent Measurements/methods , Light
2.
Mol Cell Proteomics ; 23(8): 100804, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38901673

ABSTRACT

Osmotic stress significantly hampers plant growth and crop yields, emphasizing the need for a thorough comprehension of the underlying molecular responses. Previous research has demonstrated that osmotic stress rapidly induces calcium influx and signaling, along with the activation of a specific subset of protein kinases, notably the Raf-like protein (RAF)-sucrose nonfermenting-1-related protein kinase 2 (SnRK2) kinase cascades within minutes. However, the intricate interplay between calcium signaling and the activation of RAF-SnRK2 kinase cascades remains elusive. Here, in this study, we discovered that Raf-like protein (RAF) kinases undergo hyperphosphorylation in response to osmotic shocks. Intriguingly, treatment with the calcium chelator EGTA robustly activates RAF-SnRK2 cascades, mirroring the effects of osmotic treatment. Utilizing high-throughput data-independent acquisition-based phosphoproteomics, we unveiled the global impact of EGTA on protein phosphorylation. Beyond the activation of RAFs and SnRK2s, EGTA treatment also activates mitogen-activated protein kinase cascades, Calcium-dependent protein kinases, and receptor-like protein kinases, etc. Through overlapping assays, we identified potential roles of mitogen-activated protein kinase kinase kinase kinases and receptor-like protein kinases in the osmotic stress-induced activation of RAF-SnRK2 cascades. Our findings illuminate the regulation of phosphorylation and cellular events by Ca2+ signaling, offering insights into the (exocellular) Ca2+ deprivation during early hyperosmolality sensing and signaling.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Egtazic Acid , Mannitol , Osmotic Pressure , Proteomics , Arabidopsis/metabolism , Arabidopsis/drug effects , Phosphorylation , Arabidopsis Proteins/metabolism , Proteomics/methods , Egtazic Acid/pharmacology , Egtazic Acid/analogs & derivatives , Mannitol/pharmacology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , raf Kinases/metabolism
3.
Biochem Biophys Res Commun ; 725: 150267, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38908065

ABSTRACT

Cell-to-cell transmission of α-synuclein (α-syn) pathology underlies the spread of neurodegeneration in Parkinson's disease. α-Syn secretion is an important factor in the transmission of α-syn pathology. However, it is unclear how α-syn secretion is therapeutically modulated. Here, we investigated effects of monoamine oxidase (MAO)-B inhibitor selegiline on α-syn secretion. Treatment with selegiline promoted α-syn secretion in mouse primary cortical neuron cultures, and this increase was kept under glial cell-eliminated condition by Ara-C. Selegiline-induced α-syn secretion was blocked by cytosolic Ca2+ chelator BAPTA-AM in primary neurons. Selegiline-induced α-syn secretion was retained in MAOA siRNA knockdown, whereas it was abrogated by ATG5 knockdown in SH-SY5Y cells. Selegiline increased LC3-II generation with a reduction in intracellular p62/SQSTM1 levels in primary neurons. The increase in LC3-II generation was blocked by co-treatment with BAPTA-AM in primary neurons. Additionally, fractionation experiments showed that selegiline-induced α-syn secretion occurred in non-extracellular vesicle fractions of primary neurons and SH-SY5Y cells. Collectively, these findings show that selegiline promotes neuronal autophagy involving secretion of non-exosomal α-syn via a change of cytosolic Ca2+ levels.


Subject(s)
Autophagy , Neurons , Selegiline , alpha-Synuclein , Selegiline/pharmacology , Animals , Autophagy/drug effects , alpha-Synuclein/metabolism , Neurons/drug effects , Neurons/metabolism , Mice , Monoamine Oxidase/metabolism , Humans , Calcium/metabolism , Cells, Cultured , Monoamine Oxidase Inhibitors/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Mice, Inbred C57BL , Cell Line, Tumor , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/genetics
4.
J Biol Chem ; 300(7): 107419, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815862

ABSTRACT

Extracellular secretion is an essential mechanism for α-synuclein (α-syn) proteostasis. Although it has been reported that neuronal activity affects α-syn secretion, the underlying mechanisms remain unclear. Here, we investigated the autophagic processes that regulate the physiological release of α-syn in mouse primary cortical neurons and SH-SY5Y cells. Stimulating neuronal activity with glutamate or depolarization with high KCl enhanced α-syn secretion. This glutamate-induced α-syn secretion was blocked by a mixture of NMDA receptor antagonist AP5 and AMPA receptor antagonist NBQX, as well as by cytosolic Ca2+ chelator BAPTA-AM. Additionally, mTOR inhibitor rapamycin increased α-syn and p62/SQSTM1 (p62) secretion, and this effect of rapamycin was reduced in primary cortical neurons deficient in the autophagy regulator beclin 1 (derived from BECN1+/- mice). Glutamate-induced α-syn and p62 secretion was suppressed by the knockdown of ATG5, which is required for autophagosome formation. Glutamate increased LC3-II generation and decreased intracellular p62 levels, and the increase in LC3-II levels was blocked by BAPTA-AM. Moreover, glutamate promoted co-localization of α-syn with LC3-positive puncta, but not with LAMP1-positive structures in the neuronal somas. Glutamate-induced α-syn and p62 secretion were also reduced by the knockdown of RAB8A, which is required for autophagosome fusion with the plasma membrane. Collectively, these findings suggest that stimulating neuronal activity mediates autophagic α-syn secretion in a cytosolic Ca2+-dependent manner, and autophagosomes may participate in autophagic secretion by functioning as α-syn carriers.


Subject(s)
Autophagy , Neurons , Sequestosome-1 Protein , alpha-Synuclein , Animals , Humans , Mice , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/genetics , Beclin-1/metabolism , Beclin-1/genetics , Calcium/metabolism , Cell Line, Tumor , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Glutamic Acid/metabolism , Neurons/metabolism , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Sirolimus/pharmacology
5.
Nucleic Acids Res ; 52(9): 4784-4798, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38621757

ABSTRACT

Antisense oligonucleotide (ASO) therapy is a novel therapeutic approach in which ASO specifically binds target mRNA, resulting in mRNA degradation; however, cellular uptake of ASOs remains critically low, warranting improvement. Transient receptor potential canonical (TRPC) channels regulate Ca2+ influx and are activated upon stimulation by phospholipase C-generated diacylglycerol. Herein, we report that a novel TRPC3/C6/C7 activator, L687, can induce cellular ASO uptake. L687-induced ASO uptake was enhanced in a dose- and incubation-time-dependent manner. L687 enhanced the knockdown activity of various ASOs both in vitro and in vivo. Notably, suppression of TRPC3/C6 by specific siRNAs reduced ASO uptake in A549 cells. Application of BAPTA-AM, a Ca2+ chelator, and SKF96365, a TRPC3/C6 inhibitor, suppressed Ca2+ influx via TRPC3/C6, resulting in reduced ASO uptake, thereby suggesting that Ca2+ influx via TRPC3/C6 is critical for L687-mediated increased ASO uptake. L687 also induced dextran uptake, indicating that L687 increased endocytosis. Adding ASO to L687 resulted in endosome accumulation; however, the endosomal membrane disruptor UNC7938 facilitated endosomal escape and enhanced knockdown activity. We discovered a new function for TRPC activators regarding ASO trafficking in target cells. Our findings provide an opportunity to formulate an innovative drug delivery system for the therapeutic development of ASO.


Subject(s)
Calcium , Oligonucleotides, Antisense , TRPC Cation Channels , Humans , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/metabolism , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/antagonists & inhibitors , Calcium/metabolism , A549 Cells , Animals , Mice , Imidazoles/pharmacology , TRPC6 Cation Channel/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/antagonists & inhibitors , Egtazic Acid/pharmacology , Egtazic Acid/analogs & derivatives , Endosomes/metabolism , Endosomes/drug effects , Cell Line, Tumor
6.
Int Immunopharmacol ; 131: 111916, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38522138

ABSTRACT

BACKGROUND: TRP protein is sensitive to external temperature changes, but its pathogenic mechanism in the upper airway mucosa is still unclear. OBJECTIVE: To investigate the mechanism of TRPV1and TRPA1 in regulating the secretion of inflammatory factors in nasal epithelial cells. METHODS: The expression of TRPV1 and TRPA1 in nasal mucosal epithelial cells was investigated using immunofluorescence assays. Epithelial cells were stimulated with TRPV1 and TRPA1 agonists and antagonists, and changes in Ca2+ release and inflammatory factor secretion in epithelial cells were detected. TSLP secretion stimulated with the calcium chelating agent EGTA was evaluated. The transcription factor NFAT was observed by immunofluorescence staining. RESULTS: TRPV1 and TRPA1 expression was detected in nasal epithelial cells, and Ca2+ influx was increased after stimulation with agonists. After the activation of TRPV1 and TRPA1, the gene expression of TSLP, IL-25, and IL-33 and the protein expression levels of TSLP and IL-33 were increased, and only TSLP could be inhibited by antagonists and siRNAs. After administration of EGTA, the secretion of TSLP was inhibited significantly, and the expression of the transcription factor NFAT in the nucleus was observed after activation of the TRPV1 and TRPA1 proteins in epithelial cells. CONCLUSION: Activation of TRPV1 and TRPA1 on nasal epithelial cells stimulates the generation of TSLP through the Ca2+/NFAT pathway. It also induces upregulation of IL-25 and IL-33 gene expression levels and increased levels of IL-33 protein, leading to the development of airway inflammation.


Subject(s)
Interleukin-33 , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/metabolism , Interleukin-33/metabolism , Egtazic Acid/metabolism , Gene Expression , Nasal Mucosa/metabolism , Epithelial Cells/metabolism , Transcription Factors/genetics
7.
Cell Commun Signal ; 22(1): 92, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38303059

ABSTRACT

Inherited retinal degenerations (IRDs) are a group of untreatable and commonly blinding diseases characterized by progressive photoreceptor loss. IRD pathology has been linked to an excessive activation of cyclic nucleotide-gated channels (CNGC) leading to Na+- and Ca2+-influx, subsequent activation of voltage-gated Ca2+-channels (VGCC), and further Ca2+ influx. However, a connection between excessive Ca2+ influx and photoreceptor loss has yet to be proven.Here, we used whole-retina and single-cell RNA-sequencing to compare gene expression between the rd1 mouse model for IRD and wild-type (wt) mice. Differentially expressed genes indicated links to several Ca2+-signalling related pathways. To explore these, rd1 and wt organotypic retinal explant cultures were treated with the intracellular Ca2+-chelator BAPTA-AM or inhibitors of different Ca2+-permeable channels, including CNGC, L-type VGCC, T-type VGCC, Ca2+-release-activated channel (CRAC), and Na+/Ca2+ exchanger (NCX). Moreover, we employed the novel compound NA-184 to selectively inhibit the Ca2+-dependent protease calpain-2. Effects on the retinal activity of poly(ADP-ribose) polymerase (PARP), sirtuin-type histone-deacetylase, calpains, as well as on activation of calpain-1, and - 2 were monitored, cell death was assessed via the TUNEL assay.While rd1 photoreceptor cell death was reduced by BAPTA-AM, Ca2+-channel blockers had divergent effects: While inhibition of T-type VGCC and NCX promoted survival, blocking CNGCs and CRACs did not. The treatment-related activity patterns of calpains and PARPs corresponded to the extent of cell death. Remarkably, sirtuin activity and calpain-1 activation were linked to photoreceptor protection, while calpain-2 activity was related to degeneration. In support of this finding, the calpain-2 inhibitor NA-184 protected rd1 photoreceptors.These results suggest that Ca2+ overload in rd1 photoreceptors may be triggered by T-type VGCCs and NCX. High Ca2+-levels likely suppress protective activity of calpain-1 and promote retinal degeneration via activation of calpain-2. Overall, our study details the complexity of Ca2+-signalling in photoreceptors and emphasizes the importance of targeting degenerative processes specifically to achieve a therapeutic benefit for IRDs. Video Abstract.


Subject(s)
Egtazic Acid/analogs & derivatives , Retinal Degeneration , Sirtuins , Mice , Animals , Retinal Degeneration/metabolism , Calpain/metabolism , Sodium-Calcium Exchanger , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Cell Death , Sirtuins/metabolism
8.
Int J Mol Med ; 53(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38390952

ABSTRACT

Calcium overload, a notable instigator of acute pancreatitis (AP), induces oxidative stress and an inflammatory cascade, subsequently activating both endogenous and exogenous apoptotic pathways. However, there is currently lack of available pharmaceutical interventions to alleviate AP by addressing calcium overload. In the present study, the potential clinical application of liposome nanoparticles (LNs) loaded with 1,2­bis(2­aminophenoxy)ethane­N,N,N',N'­tetraacetic acid tetrakis (acetoxymethyl ester) (BAPTA­AM), a cell­permeant calcium chelator, was investigated as a therapeutic approach for the management of AP. To establish the experimental models in vitro, AR42J cells were exposed to high glucose/sodium oleate (HGO) to induce necrosis, and in vivo, intra­ductal taurocholate (TC) infusion was used to induce AP. The findings of the present study indicated that the use of BAPTA­AM­loaded LN (BLN) effectively and rapidly eliminated excessive Ca2+ and reactive oxygen species, suppressed mononuclear macrophage activation and the release of inflammatory cytokines, and mitigated pancreatic acinar cell apoptosis and necrosis induced by HGO. Furthermore, the systemic administration of BLN demonstrated promising therapeutic potential in the rat model of AP. Notably, BLN significantly enhanced the survival rates of rats subjected to the TC challenge, increasing from 37.5 to 75%. This improvement was attributed to the restoration of pancreatic function, as indicated by improved blood biochemistry indices and alleviation of pancreatic lesions. The potential therapeutic efficacy of BLN in rescuing patients with AP is likely attributed to its capacity to inhibit oxidative stress, prevent premature activation of zymogens and downregulate the expression of TNF­α, IL­6 and cathepsin B. Thus, BLN demonstrated promising value as a novel therapeutic approach for promptly alleviating the burden of intracellular Ca2+ overload in patients with AP.


Subject(s)
Egtazic Acid/analogs & derivatives , Pancreatitis , Humans , Rats , Animals , Pancreatitis/metabolism , Liposomes/metabolism , Calcium/metabolism , Acute Disease , Acinar Cells/pathology , Necrosis/metabolism
9.
Biochim Biophys Acta Mol Cell Res ; 1871(2): 119589, 2024 02.
Article in English | MEDLINE | ID: mdl-37739271

ABSTRACT

Intracellular Ca2+ signals play a vital role in a broad range of cell biological and physiological processes in all eukaryotic cell types. Dysregulation of Ca2+ signaling has been implicated in numerous human diseases. Over the past four decades, the understanding of how cells use Ca2+ as a messenger has flourished, largely because of the development of reporters that enable visualization of Ca2+ signals in different cellular compartments, and tools that can modulate cellular Ca2+ signaling. One such tool that is frequently used is BAPTA; a fast, high-affinity Ca2+-chelating molecule. By making use of a cell-permeable acetoxymethyl ester (AM) variant, BAPTA can be readily loaded into the cytosol of cells (referred to as BAPTAi), where it is trapped and able to buffer changes in cytosolic Ca2+. Due to the ease of loading of the AM version of BAPTA, this reagent has been used in hundreds of studies to probe the role of Ca2+ signaling in specific processes. As such, for decades, researchers have almost universally attributed changes in biological processes caused by BAPTAi to the involvement of Ca2+ signaling. However, BAPTAi has often been used without any form of control, and in many cases has neither been shown to be retained in cells for the duration of experiments nor to buffer any Ca2+ signals. Moreover, increasing evidence points to off-target cellular effects of BAPTA that are clearly not related to Ca2+ chelation. Here, we briefly introduce Ca2+ signaling and the history of Ca2+ chelators and fluorescent Ca2+ indicators. We highlight Ca2+-independent effects of BAPTAi on a broad range of molecular targets and describe some of BAPTAi's impacts on cell functions that occur independently of its Ca2+-chelating properties. Finally, we propose strategies for determining whether Ca2+ chelation, the binding of other metal ions, or off-target interactions with cell components are responsible for BAPTAi's effect on a particular process and suggest some future research directions.


Subject(s)
Chelating Agents , Humans , Egtazic Acid/pharmacology , Chelating Agents/pharmacology , Cytosol
10.
Mol Biol Cell ; 35(2): ar24, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38088892

ABSTRACT

PLEKHG4B is a Cdc42-targeting guanine-nucleotide exchange factor implicated in forming epithelial cell-cell junctions. Here we explored the mechanism regulating PLEKHG4B localization. PLEKHG4B localized to the basal membrane in normal Ca2+ medium but accumulated at cell-cell junctions upon ionomycin treatment. Ionomycin-induced junctional localization of PLEKHG4B was suppressed upon disrupting its annexin-A2 (ANXA2)-binding ability. Thus, Ca2+ influx and ANXA2 binding are crucial for PLEKHG4B localization to cell-cell junctions. Treatments with low Ca2+ or BAPTA-AM (an intracellular Ca2+ chelator) suppressed PLEKHG4B localization to the basal membrane. Mutations of the phosphoinositide-binding motif in the pleckstrin homology (PH) domain of PLEKHG4B or masking of membrane phosphatidylinositol-4,5-biphosphate [PI(4,5)P2] suppressed PLEKHG4B localization to the basal membrane, indicating that basal membrane localization of PLEKHG4B requires suitable intracellular Ca2+ levels and PI(4,5)P2 binding of the PH domain. Activation of mechanosensitive ion channels (MSCs) promoted PLEKHG4B localization to cell-cell junctions, and their inhibition suppressed it. Moreover, similar to the PLEKHG4B knockdown phenotypes, inhibition of MSCs or treatment with BAPTA-AM disturbed the integrity of actin filaments at cell-cell junctions. Taken together, our results suggest that Ca2+ influx plays crucial roles in PLEKHG4B localization to cell-cell junctions and the integrity of junctional actin organization, with MSCs contributing to this process.


Subject(s)
Calcium , Egtazic Acid/analogs & derivatives , Intercellular Junctions , Calcium/metabolism , Ionomycin , Intercellular Junctions/metabolism , Actin Cytoskeleton/metabolism
11.
Biochem Biophys Res Commun ; 693: 149378, 2024 01 22.
Article in English | MEDLINE | ID: mdl-38100999

ABSTRACT

Selective calcium chelator 1,2-Bis(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid (BAPTA) is a common tool to investigate calcium signaling. However, BAPTA expresses various effects on intracellular calcium signaling, which are not related to its ability to bind Ca2+. In patch clamp experiments, we investigated calcium chelation independent effects of BAPTA on endogenous calcium-activated chloride channels ANO6 (TMEM16F) in HEK293T cells. We have found that application of BAPTA to intracellular solution led to two distinct effects on channels properties. On the one hand, application of BAPTA acutely reduced amplitude of endogenous ANO6 channels induced by 10 µM Ca2+ in single channel recordings. On the other hand, BAPTA application by itself induced ANO6 channel activity in the absence of the intracellular calcium elevation. Open channel probability was enhanced by increasing the intracellular BAPTA concentration from 0.1 to 1 and 10 mM. Another calcium chelator EGTA did not demonstrate chelation independent effects on the ANO6 activity in the same conditions. Due to off-target effects BAPTA should be used with caution when studying calcium-activated ANO6 channels.


Subject(s)
Calcium Channels , Calcium , Humans , Egtazic Acid/pharmacology , Calcium/metabolism , HEK293 Cells , Calcium Chelating Agents/pharmacology
12.
Cell Death Dis ; 14(9): 600, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37684238

ABSTRACT

Intracellular Ca2+ signals control several physiological and pathophysiological processes. The main tool to chelate intracellular Ca2+ is intracellular BAPTA (BAPTAi), usually introduced into cells as a membrane-permeant acetoxymethyl ester (BAPTA-AM). Previously, we demonstrated that BAPTAi enhanced apoptosis induced by venetoclax, a BCL-2 antagonist, in diffuse large B-cell lymphoma (DLBCL). This finding implied a novel interplay between intracellular Ca2+ signaling and anti-apoptotic BCL-2 function. Hence, we set out to identify the underlying mechanisms by which BAPTAi enhances cell death in B-cell cancers. In this study, we discovered that BAPTAi alone induced apoptosis in hematological cancer cell lines that were highly sensitive to S63845, an MCL-1 antagonist. BAPTAi provoked a rapid decline in MCL-1-protein levels by inhibiting mTORC1-driven Mcl-1 translation. These events were not a consequence of cell death, as BAX/BAK-deficient cancer cells exhibited similar downregulation of mTORC1 activity and MCL-1-protein levels. Next, we investigated how BAPTAi diminished mTORC1 activity and identified its ability to impair glycolysis by directly inhibiting 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) activity, a previously unknown effect of BAPTAi. Notably, these effects were also induced by a BAPTAi analog with low affinity for Ca2+. Consequently, our findings uncover PFKFB3 inhibition as an Ca2+-independent mechanism through which BAPTAi impairs cellular metabolism and ultimately compromises the survival of MCL-1-dependent cancer cells. These findings hold two important implications. Firstly, the direct inhibition of PFKFB3 emerges as a key regulator of mTORC1 activity and a promising target in MCL-1-dependent cancers. Secondly, cellular effects caused by BAPTAi are not necessarily related to Ca2+ signaling. Our data support the need for a reassessment of the role of Ca2+ in cellular processes when findings were based on the use of BAPTAi.


Subject(s)
Neoplasms , Phosphoric Monoester Hydrolases , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Egtazic Acid , Phosphofructokinase-2/genetics
13.
ACS Appl Mater Interfaces ; 15(34): 40163-40177, 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37603390

ABSTRACT

The bone-targeting mechanism of clinic bisphosphonate-type drugs, such as alendronate, risedronate, and ibandronate, relies on chelated calcium ions on the surface of the bone mineralized matrix for the treatment of osteoporosis. EGTA with aminocarboxyl chelating ligands can specifically chelate calcium ions. Inspired by the bone-targeting mechanism of bisphosphonates, we hypothesize that EGTA-derived carbon dots (EGTA-CDs) hold bone-targeting ability. For the target-oriented synthesis of EGTA-CDs and to endow CDs with bone targeting, we designed calcium ion chelating agents as precursors, including aminocarboxyl chelating agents (EGTA and EDTA) and bisphosphonate agents (ALN and HEDP) for the target-oriented synthesis of aminocarboxyl-derived CDs (EGTA-CDs and EDTA-CDs) and bisphosphonate-derived CDs (ALN-CDs and HEDP-CDs) with high synthetic yield. The synthetic yield of EGTA-CDs reached 87.6%. Aminocarboxyl-derived CDs and bisphosphonate-derived CDs retain the chelation ability of calcium ions and can specifically bind calcium ions. The chemical environment bone-targeting value coordination constant K and chelation sites of EGTA-CDs were 6.48 × 104 M-1 and 4.12, respectively. A novel method was established to demonstrate the bone-targeting capability of chelate-functionalized carbon dots using fluorescence quenching in a simulated bone trauma microenvironment. EGTA-CDs exhibit superior bone-targeting ability compared with other aminocarboxyl-derived CDs and bisphosphonate-derived CDs. EGTA-CDs display exceptional specificity toward calcium ions and better bone affinity than ALN-CDs, suggesting their potential as novel bone-targeting drugs. EGTA-CDs with strong calcium ion chelating ability have calcium ion affinity in simulated body fluid and bone-targeting ability in a simulated bone trauma microenvironment. These findings offer new avenues for the development of advanced bone-targeting strategies.


Subject(s)
Calcium , Etidronic Acid , Egtazic Acid , Edetic Acid , Chelating Agents/pharmacology , Diphosphonates/pharmacology , Carbon
14.
Water Environ Res ; 95(4): e10862, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37032435

ABSTRACT

The combination of UV and water-soluble Fe(III) complexes is an effective method for generating Fe(II) in situ for activating advanced oxidation processes. This study explored the potential of Fe(III)-diethylenetriaminepentaacetic acid (Fe(III)-DTPA) and Fe(III)-ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (Fe(III)-EGTA) in activating the UV/persulfate (UV/PS) for sulfamethazine removal. The initial screening showed that Fe(III)-EGTA and Fe(III)-DTPA could significantly improve the rate of sulfamethazine removal. The optimum molar ratios of persulfate to Fe(III)-DTPA and Fe(III)-EGTA were 100:1 and 100:2.5. The predicted percentage of sulfamethazine removal under the optimized conditions, obtained using response surface methodology, was ~99% for both catalysts. The pH range of 6 to 8 did not significantly affect the performance of UV/PS in the removal of sulfamethazine. The percentage sulfamethazine removal in the selected water samples was ranged from 93.6% to 99.6%, agreeing with the predicted value. The performance of both catalysts in activating UV/PS is comparable with that of the frequently used Fe(III)-EDDS. PRACTITIONERS POINTS: The potential of Fe(III)-DTPA and Fe(III)-EGTA in activating UV/persulfate (UV/PS) was explored. Fe(III)-DTPA and Fe(III)-EGTA improved the performance of UV/PS in sulfamethazine removal. Fe(III)-DTPA and Fe(III)-EGTA are effective in catalyzing UV/PS under pH 6 to 8. The performance of Fe(III)-DTPA and Fe(III)-EGTA is comparable with well-studied Fe(III)-EDDS.


Subject(s)
Sulfamethazine , Water Pollutants, Chemical , Ferric Compounds , Pentetic Acid , Egtazic Acid , Water Pollutants, Chemical/chemistry , Sulfates/chemistry , Oxidation-Reduction , Water
15.
Cell Calcium ; 112: 102738, 2023 06.
Article in English | MEDLINE | ID: mdl-37060673

ABSTRACT

In platelets, elevated cytosolic Ca2+ is a crucial second messenger, involved in most functional responses, including shape change, secretion, aggregation and procoagulant activity. The platelet Ca2+ response consists of Ca2+ mobilization from endoplasmic reticulum stores, complemented with store-operated or receptor-operated Ca2+ entry pathways. Several channels can contribute to the Ca2+ entry, but their relative contribution is unclear upon stimulation of ITAM-linked receptors such as glycoprotein VI (GPVI) and G-protein coupled receptors such as the protease-activated receptors (PAR) for thrombin. We employed a 96-well plate high-throughput assay with Fura-2-loaded human platelets to perform parallel [Ca2+]i measurements in the presence of EGTA or CaCl2. Per agonist condition, this resulted in sets of EGTA, CaCl2 and Ca2+ entry ratio curves, defined by six parameters, reflecting different Ca2+ ion fluxes. We report that threshold stimulation of GPVI or PAR, with a variable contribution of secondary mediators, induces a maximal Ca2+ entry ratio of 3-7. Strikingly, in combination with Ca2+-ATPase inhibition by thapsigargin, the maximal Ca2+ entry ratio increased to 400 (GPVI) or 40 (PAR), pointing to a strong receptor-dependent enhancement of store-operated Ca2+ entry. By pharmacological blockage of specific Ca2+ channels in platelets, we found that, regardless of GPVI or PAR stimulation, the Ca2+ entry ratio was strongest affected by inhibition of ORAI1 (2-APB, Synta66) > Na+/Ca2+ exchange (NCE) > P2×1 (only initial). In contrast, inhibition of TRPC6, Piezo1/2 or STIM1 was without effect. Together, these data reveal ORAI1 and NCE as dominating Ca2+ carriers regulating GPVI- and PAR-induced Ca2+ entry in human platelets.


Subject(s)
Blood Platelets , Calcium Channels , Humans , Blood Platelets/metabolism , Calcium Channels/metabolism , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/pharmacology , Calcium Chloride/pharmacology , Egtazic Acid/metabolism , Calcium Signaling , Receptors, G-Protein-Coupled/metabolism , Calcium/metabolism , Stromal Interaction Molecule 1/metabolism , ORAI1 Protein/metabolism , Ion Channels/metabolism
16.
Acta Physiol (Oxf) ; 237(3): e13922, 2023 03.
Article in English | MEDLINE | ID: mdl-36599422

ABSTRACT

AIM: Gold nanoparticles are widely used for biomedical applications, but the precise molecular mechanism of their interaction with cellular structures is still unclear. Assuming that intracellular calcium fluctuations associated with surface plasmon-induced calcium entry could modulate the activity of potassium channels, we studied the effect of 5 nm gold nanoparticles on calcium-dependent potassium channels and associated calcium signaling in freshly isolated rat pulmonary artery smooth muscle cells and cultured hippocampal neurons. METHODS: Outward potassium currents were recorded using patch-clamp techniques. Changes in intracellular calcium concentration were measured using the high affinity Ca2+ fluorescent indicator fluo-3 and laser confocal microscope. RESULTS: In pulmonary artery smooth muscle cells, plasmonic gold nanoparticles increased the amplitude of currents via large-conductance Ca2+ -activated potassium channels, which was potentiated by green laser irradiation near plasmon resonance wavelength (532 nm). Buffering of intracellular free calcium with ethylene glycol-bis-N,N,N',N'-tetraacetic acid (EGTA) abolished these effects. Furthermore, using confocal laser microscopy it was found that application of gold nanoparticles caused oscillations of intracellular calcium concentration that were decreasing in amplitude with time. In cultured hippocampal neurons gold nanoparticles inhibited the effect of EGTA slowing down the decline of the BKCa current while partially restoring the amplitude of the slow after hyperpolarizing currents. CONCLUSION: We conclude that fluctuations in intracellular calcium can modulate plasmonic gold nanoparticles-induced gating of BKCa channels in smooth muscle cells and neurons through an indirect mechanism, probably involving the interaction of plasmon resonance with calcium-permeable ion channels, which leads to a change in intracellular calcium level.


Subject(s)
Hippocampus , Metal Nanoparticles , Myocytes, Smooth Muscle , Potassium Channels , Animals , Rats , Calcium/metabolism , Egtazic Acid , Gold/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Metal Nanoparticles/therapeutic use , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Pulmonary Artery/metabolism
17.
Int J Phytoremediation ; 25(9): 1106-1115, 2023.
Article in English | MEDLINE | ID: mdl-36264021

ABSTRACT

This study focused to enhance the cadmium (Cd) phytoextraction efficiency in Solanum nigrum by applying four biodegradable chelants (10 mM)-ethylene glycol tetraacetic acid (EGTA), ethylenediamine disuccinate (EDDS), nitrilotriacetic acid (NTA), and citric acid (CA), when grown in Cd-spiked soil (12 and 48 mg kg-1). Plant height, dry biomass, photosynthetic traits, and metal accumulation varied significantly with Cd and chelant treatments. Cadmium-toxicity resulted in reduction of plant growth and photosynthetic physiology, whereas chelant supplementation alleviated the toxic effect of Cd and increased its accumulation. Tolerance index value increased with addition of chelants in the order: EGTA (1.57-1.63) >EDDS (1.39-1.58) >NTA (1.14-1.50) >CA (1-1.22) compared with Cd (0.46-1.08). Transfer coefficient of root increased with supplementation of EGTA (3.40-3.85), EDDS (3.10-3.40), NTA (2.60-2.90), and CA (1.85-2.29), over Cd-alone (1.61-1.63). Similarly, translocation factor was also increased upon addition of EGTA (0.52-0.73), EDDS (0.35-0.81), NTA (0.38-0.75), and CA (0.53-0.54), compared with Cd-alone (0.36-0.59). Maximum Cd removal (67.67% at Cd12 and 36.05% at Cd48) was observed with supplementation of EGTA. The study concludes that the supplementation of EGTA and EDDS with S. nigrum can be employed as an efficient and environmentally safe technique for reclamation of Cd-contaminated soils.


Apart from the selection of a good hyperaccumulator, the choice of chelant (biodegradable/non-biodegradable) is an important aspect for the successful phytoextraction of metals from contaminated soil. We reported for the first time the potential of ethylene glycol tetraacetic acid (EGTA; a biodegradable chelant) in enhancing Cd phytoextraction by Solanum nigrum. Comparative appraisal of metal extraction efficiency of biodegradable chelants at low (12 mg kg−1) and high (48 mg kg−1) Cd dose depicted that EGTA performed better than EDDS, NTA, and CA (other biodegradable chelants). EGTA supplementation did not induce toxicity in plants; rather it improved metal accumulation, morphology, and photosynthetic physiology.


Subject(s)
Soil Pollutants , Solanum nigrum , Cadmium , Chelating Agents/pharmacology , Egtazic Acid , Biodegradation, Environmental , Soil Pollutants/analysis , Nitrilotriacetic Acid , Soil , Citric Acid
18.
Cell Mol Neurobiol ; 43(5): 2257-2271, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36369494

ABSTRACT

Acetylcholine can excite neurons by suppressing M-type (KCNQ) potassium channels. This effect is mediated by M1 muscarinic receptors coupled to the Gq protein. Although PIP2 depletion and PKC activation have been strongly suggested to contribute to muscarinic inhibition of M currents (IM), direct evidence is lacking. We investigated the mechanism involved in muscarinic inhibition of IM with Ca2+ measurement and electrophysiological studies in both neuronal (rat sympathetic neurons) and heterologous (HEK cells expressing KCNQ2/KCNQ3) preparations. We found that muscarinic inhibition of IM was not blocked either by PIP2 or by calphostin C, a PKC inhibitor. We then examined whether muscarinic inhibition of IM uses multiple signaling pathways by blocking both PIP2 depletion and PKC activation. This maneuver, however, did not block muscarinic inhibition of IM. Additionally, muscarinic inhibition of IM was not prevented either by sequestering of G-protein ßγ subunits from Gα-transducin or anti-Gßγ antibody or by preventing intracellular trafficking of channel proteins with blebbistatin, a class-II myosin inhibitor. Finally, we re-examined the role of Ca2+ signals in muscarinic inhibition of IM. Ca2+ measurements showed that muscarinic stimulation increased intracellular Ca2+ and was comparable to the Ca2+ mobilizing effect of bradykinin. Accordingly, 20-mM of BAPTA significantly suppressed muscarinic inhibition of IM. In contrast, muscarinic inhibition of IM was completely insensitive to 20-mM EGTA. Taken together, these data suggest a role of Ca2+ signaling in muscarinic modulation of IM. The differential effects of EGTA and BAPTA imply that Ca2+ microdomains or spatially local Ca2+ signals contribute to inhibition of IM.


Subject(s)
Neurons , Signal Transduction , Rats , Animals , Egtazic Acid/metabolism , Egtazic Acid/pharmacology , Neurons/metabolism , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology
19.
Biotechnol Lett ; 45(2): 199-207, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36504267

ABSTRACT

Reactive Oxygen Species (ROS) play an important role in oxidative stress and are related to the lipid accumulation in microalgae. Nicotinamide Adenine Dinucleotide Phosphate (NADPH) oxidase can oxidize O2 to O2- ultimately. However, the function of NADPH oxidase and its contribution to the production of the intracellular total ROS are still unclear. In this study, the function of NADPH oxidase in Chlorella pyrenoidosa (C. pyrenoidosa) was investigated by adding activators Ca2+ and NADPH and inhibitors EGTA, LaCl3, DPI and BAPTA of NADPH oxidase. The results show that the addition of activators of Ca2+ or NADPH significantly increased the intracellular concentrations of ROS molecules (H2O2, O2-, and OH·) in C. pyrenoidosa. Moreover, the intracellular ROS level was higher under the nitrogen-deficient and phosphorus-deficient conditions than in control condition, but the addition of the inhibitors (EGTA, LaCl3, DPI, and BAPTA) of NADPH oxidase significantly reduced the intracellular concentrations of H2O2, O2-, and OH·. The study shows that NADPH oxidase actively participated in the production of intracellular ROS in C. pyrenoidosa, demonstrating that NADPH oxidase was another important element in the production of intracellular ROS in addition to mitochondria, chloroplasts and lysozymes in microalgae.


Subject(s)
Chlorella , Reactive Oxygen Species , Egtazic Acid , Hydrogen Peroxide/pharmacology , NADP , NADPH Oxidases
20.
Protoplasma ; 260(1): 299-306, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35676506

ABSTRACT

The hydraulic resistance (the reciprocal of the hydraulic conductivity Lp) Lp-1 was measured in cells of Chara corallina by the method of transcellular osmosis. Treatment of cells with 100 mM KCl decreased Lp-1 significantly. Subsequent treatment of the cells with 70 mM CaCl2 recovered the decreased Lp-1 to the original value. To know whether K+ or Ca2+/Mg2+ acts on the cell wall and/or the membrane, the hydraulic resistances of the cell wall (Lpw-1) and that of the membrane (Lpm-1) were determined in one and the same cell. For this, a pair of cells (twin cells) were made from an internodal cell, one used for measurement of Lp-1 and the other used for the measurement of Lpw-1. From Lp-1 and Lpw-1, Lpm-1 was calculated. Both Lp-1 and Lpw-1 were decreased by K+, while Lpm-1 was not affected by K+. The same result was obtained with 5 mM EGTA. Lpw-1 was decreased more than it was by KCl but Lpm-1 remained constant after EGTA treatment. The recovery of the K+-decreased Lp-1 with Ca2+ can be explained exclusively by the recovery of Lpw-1 with Ca2+. The Ca2+ recovery of Lpw-1 was observed in the intact cell wall but not in the cell wall tube isolated from an internodal cell. The different response to Ca2+ between the intact cell wall and the isolated cell wall was discussed in relation to the tension in the cell wall which may be an important factor for the ionic regulation of hydraulic conductivity.


Subject(s)
Calcium , Chara , Calcium/metabolism , Chara/physiology , Egtazic Acid/metabolism , Cell Wall/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL