Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 336
Filter
1.
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
2.
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
3.
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
4.
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
5.
J Transl Med ; 20(1): 561, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36463203

ABSTRACT

BACKGROUND: Destruction of articular cartilage and bone is the main cause of joint dysfunction in rheumatoid arthritis (RA). Acid-sensing ion channel 1a (ASIC1a) is a key molecule that mediates the destruction of RA articular cartilage. Estrogen has been proven to have a protective effect against articular cartilage damage, however, the underlying mechanisms remain unclear. METHODS: We treated rat articular chondrocytes with an acidic environment, analyzed the expression levels of mitochondrial stress protein HSP10, ClpP, LONP1 by q-PCR and immunofluorescence staining. Transmission electron microscopy was used to analyze the mitochondrial morphological changes. Laser confocal microscopy was used to analyze the Ca2+, mitochondrial membrane potential (Δψm) and reactive oxygen species (ROS) level. Moreover, ASIC1a specific inhibitor Psalmotoxin 1 (Pctx-1) and Ethylene Glycol Tetraacetic Acid (EGTA) were used to observe whether acid stimulation damage mitochondrial function through Ca2+ influx mediated by ASIC1a and whether pretreatment with estrogen could counteract these phenomena. Furthermore, the ovariectomized (OVX) adjuvant arthritis (AA) rat model was treated with estrogen to explore the effect of estrogen on disease progression. RESULTS: Our results indicated that HSP10, ClpP, LONP1 protein and mRNA expression and mitochondrial ROS level were elevated in acid-stimulated chondrocytes. Moreover, acid stimulation decreased mitochondrial membrane potential and damaged mitochondrial structure of chondrocytes. Furthermore, ASIC1a specific inhibitor PcTx-1 and EGTA inhibited acid-induced mitochondrial abnormalities. In addition, estrogen could protect acid-stimulated induced mitochondrial stress by regulating the activity of ASIC1a in rat chondrocytes and protects cartilage damage in OVX AA rat. CONCLUSIONS: Extracellular acidification induces mitochondrial stress by activating ASIC1a, leading to the damage of rat articular chondrocytes. Estrogen antagonizes acidosis-induced joint damage by inhibiting ASIC1a activity. Our study provides new insights into the protective effect and mechanism of action of estrogen in RA.


Subject(s)
Acid Sensing Ion Channels , Arthritis, Rheumatoid , Chondrocytes , Estrogens , Mitochondria , Animals , Rats , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Arthritis, Experimental , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Chondrocytes/drug effects , Chondrocytes/metabolism , Egtazic Acid/metabolism , Egtazic Acid/toxicity , Estrogens/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Reactive Oxygen Species , Cartilage, Articular/drug effects , Cartilage, Articular/pathology
6.
Zhongguo Zhong Yao Za Zhi ; 47(21): 5900-5907, 2022 Nov.
Article in Chinese | MEDLINE | ID: mdl-36472009

ABSTRACT

This study was designed to determine the inhibitory effect of astragaloside Ⅳ(AS-Ⅳ), a principal bioactive component extracted from the Chinese medicinal Astragali Radix, on the inflammatory response of vascular endothelial cells induced by angiotensin Ⅱ(Ang Ⅱ), the most major pathogenic factor for cardiovascular diseases, and to clarify the role of calcium(Ca~(2+))/phosphatidylinosi-tol-3-kinase(PI3K)/protein kinase B(Akt)/endothelial nitric oxide synthase(eNOS)/nitric oxide(NO) pathway in the process. To be specific, human umbilical vein endothelial cells(HUVECs) were cultured in the presence of AS-Ⅳ with or without the specific inhibitor of NO synthase(NG-monomethyl-L-arginine, L-NMMA), inhibitor of PI3K/Akt signaling pathway(LY294002), or Ca~(2+)-chelating agent(ethylene glycol tetraacetic acid, EGTA) prior to Ang Ⅱ stimulation. The inhibitory effect of AS-Ⅳ on Ang Ⅱ-induced inflammatory response and the involved mechanism was determined with enzyme-linked immunosorbent assay(ELISA), cell-based ELISA assay, Western blot, and monocyte adhesion assay which determined the fluorescently labeled human monocytic cell line(THP-1) adhered to Ang Ⅱ-stimulated endothelial cells. AS-Ⅳ increased the production of NO by HUVECs in a dose-and time-dependent manner(P<0.05) and raised the level of phosphorylated eNOS(P<0.05). The above AS-Ⅳ-induced changes were abolished by pretreatment with L-NMMA, LY294002, or EGTA. Compared with the control group, Ang Ⅱ obviously enhanced the production and release of cytokines(tumor necrosis factor-α, interleukin-6), chemokines(monocyte chemoattractant protein-1) and adhesion molecules(intercellular adhesion molecule-1, vascular cellular adhesion molecule-1), and the number of monocytes adhered to HUVECs(P<0.05), which were accompanied by the enhanced levels of phosphorylated inhibitor of nuclear factor-κBα protein and activities of nuclear factor-κB(NF-κB)(P<0.05). This study also demonstrated that Ang Ⅱ-induced inflammatory response was inhibited by pretreatment with AS-Ⅳ(P<0.05). In addition, the inhibitory effect of AS-Ⅳ was abrogated by pretreatment with L-NMMA, LY294002, or EGTA(P<0.05). This study provides a direct link between AS-Ⅳ and Ca~(2+)/PI3K/Akt/eNOS/NO pathway in AS-Ⅳ-mediated anti-inflammatory actions in endothelial cells exposed to Ang Ⅱ. The results indicate that AS-Ⅳ attenuates endothelial cell-mediated inflammatory response induced by Ang Ⅱ via the activation of Ca~(2+)/PI3K/Akt/eNOS/NO signaling pathway.


Subject(s)
Angiotensin II , Proto-Oncogene Proteins c-akt , Humans , Angiotensin II/metabolism , Angiotensin II/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , omega-N-Methylarginine/metabolism , omega-N-Methylarginine/pharmacology , Egtazic Acid/metabolism , Egtazic Acid/pharmacology , Human Umbilical Vein Endothelial Cells , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/metabolism , Cells, Cultured
7.
PeerJ ; 10: e14574, 2022.
Article in English | MEDLINE | ID: mdl-36570013

ABSTRACT

Fruit peel creasing is a serious pre-harvest physiological disorder in citrus, influencing fruit quality, storage, and yield. Four- and eight-year-old 'Hongjiang' oranges grafted onto Canton lemon rootstocks were treated with calcium and calcium inhibitors, respectively, to study the effects of different treatments on fruit creasing rate, mechanical properties of the peel, cell wall metabolism enzyme activities, and the expression of related genes. Foliar application of 0.5% calcium nitrate significantly reduced the fruit creasing rate, while treatment with EGTA and LaCl3, inhibitors of calcium uptake, increased the fruit creasing rate; But the effect of calcium nitrate treatment on changing the mechanical properties of pericarp and inhibiting the activity of hydrolase (PG, Cx and PE) was not very significant. Furthermore, it was observed that the expression levels of genes (PG, Cx, and PE) encoding cell wall-degrading enzymes were significantly lower in the normal fruit peel than in the creased fruit peel. Meanwhile, the expression levels of PG, Cx, and PE were higher in the peel of shaded fruit than in the peel of exposed fruit. During the high incidence period of fruit creasing, calcium nitrate treatment down-regulated the expression of PG, Cx, and PE, while EGTA treatment up-regulated the expression of these genes. In conclusion, foliar spraying of calcium nitrate at the fruit rapid enlargement stage can increase the Ca content in the peel of 'Hongjiang' orange and significantly suppress the expression of cell wall degrading enzymes genes (PG, PE and Cx) in 'Hongjiang' orange peel during the high occurrence period of fruit creasing, resulting in reducing the occurrence of fruit creasing and cracking.


Subject(s)
Citrus sinensis , Citrus , Citrus/genetics , Fruit/genetics , Egtazic Acid/metabolism , Citrus sinensis/genetics , Cell Wall/genetics
8.
Sci Rep ; 12(1): 22407, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36575295

ABSTRACT

Synaptotagmin-1 is a vesicular protein and Ca2+ sensor for Ca2+-dependent exocytosis. Ca2+ induces synaptotagmin-1 binding to its own vesicle membrane, called the cis-interaction, thus preventing the trans-interaction of synaptotagmin-1 to the plasma membrane. However, the electrostatic regulation of the cis- and trans-membrane interaction of synaptotagmin-1 was poorly understood in different Ca2+-buffering conditions. Here we provide an assay to monitor the cis- and trans-membrane interactions of synaptotagmin-1 by using native purified vesicles and the plasma membrane-mimicking liposomes (PM-liposomes). Both ATP and EGTA similarly reverse the cis-membrane interaction of synaptotagmin-1 in free [Ca2+] of 10-100 µM. High PIP2 concentrations in the PM-liposomes reduce the Hill coefficient of vesicle fusion and synaptotagmin-1 membrane binding; this observation suggests that local PIP2 concentrations control the Ca2+-cooperativity of synaptotagmin-1. Our data provide evidence that Ca2+ chelators, including EGTA and polyphosphate anions such as ATP, ADP, and AMP, electrostatically reverse the cis-interaction of synaptotagmin-1.


Subject(s)
Liposomes , Synaptotagmin I , Liposomes/metabolism , Static Electricity , Egtazic Acid/metabolism , Synaptotagmin I/metabolism , Cell Membrane/metabolism , Membrane Fusion/physiology , Exocytosis/physiology , Adenosine Triphosphate/metabolism , Calcium/metabolism , Synaptotagmins/metabolism , SNARE Proteins/metabolism
9.
J Insect Physiol ; 143: 104453, 2022.
Article in English | MEDLINE | ID: mdl-36341969

ABSTRACT

Insect nephrocytes are ultrafiltration cells that remove circulating proteins and exogenous toxins from the haemolymph. Experimental disruption of nephrocyte development or function leads to systemic impairment of insect physiology as evidenced by cardiomyopathy, chronic activation of immune signalling and shortening of lifespan. The genetic and structural basis of the nephrocyte's ultrafiltration mechanism is conserved between arthropods and mammals, making them an attractive model for studying human renal function and systemic clearance mechanisms in general. Although dynamic changes to intracellular calcium are fundamental to the function of many cell types, there are currently no studies of intracellular calcium signalling in nephrocytes. In this work we aimed to characterise calcium signalling in the pericardial nephrocytes of Drosophila melanogaster. To achieve this, a genetically encoded calcium reporter (GCaMP6) was expressed in nephrocytes to monitor intracellular calcium both in vivo within larvae and in vitro within dissected adults. Larval nephrocytes exhibited stochastically timed calcium waves. A calcium signal could be initiated in preparations of adult nephrocytes and abolished by EGTA, or the store operated calcium entry (SOCE) blocker 2-APB, as well as RNAi mediated knockdown of the SOCE genes Stim and Orai. Neither the presence of calcium-free buffer nor EGTA affected the binding of the endocytic cargo albumin to nephrocytes but they did impair the subsequent accumulation of albumin within nephrocytes. Pre-treatment with EGTA, calcium-free buffer or 2-APB led to significantly reduced albumin binding. Knock-down of Stim and Orai was non-lethal, caused an increase to nephrocyte size and reduced albumin binding, reduced the abundance of the endocytic cargo receptor Amnionless and disrupted the localisation of Dumbfounded at the filtration slit diaphragm. These data indicate that pericardial nephrocytes exhibit stochastically timed calcium waves in vivo and that SOCE mediates the localisation of the endocytic co-receptor Amnionless. Identifying the signals both up and downstream of SOCE may highlight mechanisms relevant to the renal and excretory functions of a broad range of species, including humans.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Albumins/metabolism , Calcium Signaling , Drosophila melanogaster/genetics , Drosophila Proteins/metabolism , Egtazic Acid/metabolism , Endocytosis , Larva/metabolism , Mammals/metabolism
10.
Plant Cell Rep ; 41(4): 1043-1057, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35190883

ABSTRACT

KEY MESSAGE: After cryopreservation, the Ca2+ content increased, which affected the intracellular ROS content, then participated in the occurrence of programmed cell death in pollen. Programmed cell death (PCD) is one of the reasons for the decline in pollen viability after cryopreservation. However, the role of calcium ions (Ca2+) in PCD during pollen cryopreservation has not been revealed in the existing studies. In this study, Paeonia lactiflora 'Fen Yu Nu' pollen was used as the research material for investigating the effects of Ca2+ changes on PCD indices and reactive oxygen species (ROS) during pollen cryopreservation. The results showed that after cryopreservation, with the decrease of pollen viability, the Ca2+ content significantly increased. The regulation of Ca2+ content had a significant effect on PCD indices, which showed that the Ca2+ carrier A23187 accelerated the decrease of mitochondrial membrane potential level and increased the activity of caspase-3-like and caspase-9-like proteases and the apoptosis rate. The expression levels of partial pro-PCD genes were upregulated, the anti-PCD gene BI-1 was downregulated, and the addition of Ca2+-chelating agent EGTA had the opposite effect. The addition of the Ca2+ carrier A23187 after cryopreservation significantly increased the ROS content of pollen, the addition of the Ca2+-chelating agent EGTA had the opposite effect, and Ca2+ regulators also had significant effects on the contents of ROS production and clearance-related substances. Ca2+ affected intracellular ROS content by acting on the ROS production and clearance system during the cryopreservation of pollen and is thus involved in the occurrence of PCD.


Subject(s)
Apoptosis , Pollen , Calcimycin/metabolism , Calcimycin/pharmacology , Chelating Agents/pharmacology , Cryopreservation/methods , Egtazic Acid/metabolism , Egtazic Acid/pharmacology , Pollen/genetics , Reactive Oxygen Species/metabolism
11.
Oxid Med Cell Longev ; 2022: 8287633, 2022.
Article in English | MEDLINE | ID: mdl-36600949

ABSTRACT

Calcium homeostasis plays a vital role in protecting against Alzheimer's disease (AD). In this study, amyloid-ß (Aß)-induced C. elegans models of AD were used to elucidate the mechanisms underlying calcium homeostasis in AD. Calcium acetate increased the intracellular calcium content, exacerbated Aß 1-42 aggregation, which is closely associated with oxidative stress, aggravated neuronal degeneration and dysfunction, and shortened the lifespan of the C. elegans models. Ethylene glycol tetraacetic acid (EGTA) and nimodipine were used to decrease the intracellular calcium content. Both EGTA and nimodipine showed remarkable inhibitory effects on Aß 1-42 aggregations by increasing oxidative stress resistance. Moreover, both compounds significantly delayed the onset of Aß-induced paralysis, rescued memory deficits, ameliorated behavioral dysfunction, decreased the vulnerability of two major (GABAergic and dopaminergic) neurons and synapses, and extended the lifespan of the C. elegans AD models. Furthermore, RNA sequencing of nimodipine-treated worms revealed numerous downstream differentially expressed genes related to calcium signaling. Nimodipine-induced inhibition of selective voltage-gated calcium channels was shown to activate other calcium channels of the plasma membrane (clhm-1) and endoplasmic reticulum (unc-68), in addition to sodium-calcium exchanger channels (ncx-1). These channels collaborated to activate downstream events to resist oxidative stress through glutathione S-transferase activity mediated by HPGD and skn-1, as verified by RNA interference. These results may be applied for the treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Caenorhabditis elegans Proteins , Animals , Alzheimer Disease/metabolism , Caenorhabditis elegans , Calcium/metabolism , Nimodipine/pharmacology , Nimodipine/therapeutic use , Egtazic Acid/metabolism , Egtazic Acid/pharmacology , Oxidative Stress , Amyloid beta-Peptides/metabolism , Calcium Channels/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Caenorhabditis elegans Proteins/metabolism
12.
Sci Rep ; 11(1): 7486, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33820929

ABSTRACT

Fetuin-A is an inhibitor of ectopic calcification that is expressed mainly in hepatocytes and is secreted into the circulation after posttranslational processing, including glycosylation and phosphorylation. The molecular weight (MW) of fully modified fetuin-A (FM-fetuin-A) is approximately 60 kDa in an immunoblot, which is much higher than the estimated MW by amino acid sequence. Under conditions of calcification stress such as advanced stage chronic kidney disease, fetuin-A prevents calcification by forming colloidal complexes, which are referred to as calciprotein particles (CPP). Since the significance of CPP in this process is unclear, we investigated the effect of synthetic secondary CPP on the level of FM-fetuin-A in HepG2 cells. Secondary CPP increased the level of FM-fetuin-A in dose- and time-dependent manners, but did not affect expression of mRNA for fetuin-A. Treatment with O- and/or N-glycosidase caused a shift of the 60 kDa band of FM-fetuin-A to a lower MW. Preincubation with brefeldin A, an inhibitor of transport of newly synthesized proteins from the endoplasmic reticulum to the Golgi apparatus, completely blocked the secondary CPP-induced increase in FM-fetuin-A. Treatment with BAPTA-AM, an intracellular calcium chelating agent, also inhibited the CPP-induced increase in the FM-fetuin-A level. Secondary CPP accelerate posttranslational processing of fetuin-A in HepG2 cells.


Subject(s)
Calcinosis/metabolism , Protein Processing, Post-Translational , alpha-2-HS-Glycoprotein/metabolism , Brefeldin A/pharmacology , Calcium/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Glycoside Hydrolases/metabolism , Glycosylation , Hep G2 Cells , Humans , Phosphorylation , Phosphoserine/metabolism , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , alpha-2-HS-Glycoprotein/genetics
13.
Mol Vis ; 26: 780-788, 2020.
Article in English | MEDLINE | ID: mdl-33311973

ABSTRACT

Purpose: Vasoactive intestinal peptide (VIP) is an important regulator of lacrimal gland (LG) function although the effect of VIP on ductal fluid secretion is unknown. Therefore, the aim of the present study was to investigate the role of VIP in the regulation of fluid secretion of isolated LG ducts and to analyze the underlying intracellular mechanisms. Methods: LGs from wild-type (WT) and cystic fibrosis transmembrane conductance regulator (CFTR) knockout (KO) mice were used. Immunofluorescence was applied to confirm the presence of VIP receptors termed VPAC1 and VPAC2 in LG duct cells. Ductal fluid secretion evoked by VIP (100 nM) was measured in isolated ducts using videomicroscopy. Intracellular Ca2+ signaling underlying VIP stimulation was investigated with microfluorometry. Results: VIP stimulation resulted in a robust and continuous fluid secretory response in isolated duct segments originated from WT mice. In contrast, CFTR KO ducts exhibited only a weak pulse-like secretion. A small but statistically significant increase was detected in the intracellular Ca2+ level [Ca2+]i during VIP stimulation in the WT and in CFTR KO ducts. VIP-evoked changes in [Ca2+]i did not differ considerably between the WT and CFTR KO ducts. Conclusions: These results suggest the importance of VIP in the regulation of ductal fluid secretion and the determining role of the adenylyl cyclase-cAMP-CFTR route in this process.


Subject(s)
Lacrimal Apparatus/metabolism , Tears/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Carbachol/pharmacology , Chelating Agents/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Intracellular Space/metabolism , Mice, Knockout , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism
14.
Cell Calcium ; 90: 102248, 2020 09.
Article in English | MEDLINE | ID: mdl-32629299

ABSTRACT

Human platelets use a rise in cytosolic Ca2+ concentration to activate all stages of thrombus formation, however, how they are able to decode cytosolic Ca2+ signals to trigger each of these independently is unknown. Other cells create local Ca2+ signals to activate Ca2+-sensitive effectors specifically localised to these subcellular regions. However, no previous study has demonstrated that agonist-stimulated human platelets can generate a local cytosolic Ca2+ signal. Platelets possess a structure called the membrane complex (MC) where the main intracellular calcium store, the dense tubular system (DTS), is coupled tightly to an invaginated portion of the plasma membrane called the open canalicular system (OCS). Here we hypothesised that human platelets use a Ca2+ nanodomain created within the MC to control the earliest phases of platelet activation. Dimethyl-BAPTA-loaded human platelets were stimulated with thrombin in the absence of extracellular Ca2+ to isolate a cytosolic Ca2+ nanodomain created by Ca2+ release from the DTS. In the absence of any detectable rise in global cytosolic Ca2+ concentration, thrombin stimulation triggered Na+/Ca2+ exchanger (NCX)-dependent Ca2+ removal into the extracellular space, as well as Ca2+-dependent shape change in the absence of platelet aggregation. The NCX-mediated Ca2+ removal was dependent on the normal localisation of the DTS, and immunofluorescent staining of NCX3 demonstrated its localisation to the OCS, consistent with this Ca2+ nanodomain being formed within the MC. These results demonstrated that human platelets possess a functional Ca2+ nanodomain contained within the MC that can control shape change independently of platelet aggregation.


Subject(s)
Blood Platelets/cytology , Blood Platelets/metabolism , Calcium/metabolism , Cell Shape , Cytosol/metabolism , Nanoparticles/chemistry , Platelet Aggregation , Blood Platelets/drug effects , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Shape/drug effects , Cytosol/drug effects , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Nicergoline/pharmacology , Platelet Aggregation/drug effects , Sodium-Calcium Exchanger/metabolism , Subcellular Fractions/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology , Thrombin/pharmacology
15.
Life Sci ; 253: 117675, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32360621

ABSTRACT

AIMS: Gliomas are responsible for the majority of deaths from primary brain tumours. Sevoflurane showed inhibition effects on the tumor progression in vitro. However, whether sevoflurane could affect the stemness of glioma stem cells (GSCs) and the potential molecular mechanism have not been well elucidated. MAIN METHODS: Effects of sevoflurane on cell viability, proliferation and invasion ability of glioma cells as well as tumor growth in vivo were assessed. Sphere formation assay was performed to evaluate the effect of sevoflurane on the stemness of GSCs. Effects of sevoflurane on mitochondrial function was evaluated by intracellular/mitochondrial reactive oxygen species (ROS) level and mitochondrial membrane potential. Expression levels of proliferation-related proteins, stemness markers and proteins in CaMKII/JNK cascade were measured by Western blot. KEY FINDINGS: Sevoflurane inhibited the viability, proliferation and invasion ability of glioma cells (U87MG and U373MG). Western blot showed that sevoflurane decreased the expression levels of proliferation and invasion-related proteins. Sphere formation ability of GSCs, expression levels of stemness markers and mitochondrial function were significantly suppressed by sevoflurane. Moreover, sevoflurane treatment significantly increased the Ca2+ concentration and stimulated phosphorylation of CaMKII, JNK and IRS1. Ca2+ chelator BAPTA-AM combined with sevoflurane synergistically inhibited colony forming ability and the expression levels of proliferation-related proteins and stemness markers. In addition, the in vivo study further confirmed that sevoflurane inhibited tumor growth via Ca2+-dependent CaMKII/JNK cascade. SIGNIFICANCE: The present study demonstrated that sevoflurane inhibited glioma tumorigenesis and modulated the cancer stem cell-like properties and mitochondrial membrane potential via activation of Ca2+-dependent CaMKII/JNK cascade.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , MAP Kinase Kinase 4/metabolism , Membrane Potential, Mitochondrial/drug effects , Neoplastic Stem Cells/drug effects , Sevoflurane/metabolism , Sevoflurane/pharmacology , Animals , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Chelating Agents/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Glioma , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction
16.
Cell Calcium ; 86: 102134, 2020 03.
Article in English | MEDLINE | ID: mdl-31838437

ABSTRACT

During myogenesis, a long splice variant of STIM1, called STIM1L is getting expressed, while the level of STIM1 remains constant. Previous work demonstrated that STIM1L is more efficient in eliciting store-operated Ca2+ entry (SOCE), but no current analysis of the channel(s) activated by this new STIM1L isoform was performed until now. In this study, we investigate the ionic channel(s) activated by STIM1L and whether differences exist between the two STIM1 isoforms, using HEK-293 T cells as a model system. Our data show that STIM1 and STIM1L activate Orai1 channel but also the endogenously expressed TRPC1. The channel activation occurs in two steps, with first Orai1 activation followed, in a subset of cells, by TRPC1 opening. Remarkably, STIM1L more frequently activates TRPC1 and preferentially interacts with TRPC1. In low intracellular Ca2+ buffering condition, the frequency of TRPC1 opening increases significantly, strongly suggesting a Ca2+-dependent channel activation. The ability of STIM1L to open Orai1 appears decreased compared to STIM1, which might be explained by its stronger propensity towards TRPC1. Indeed, increasing the amount of STIM1L results in an enhanced Orai1 current. The role of endogenous TRPC1 in STIM1- and STIM1L-induced SOCE was confirmed by Ca2+ imaging experiments. Overall, our findings provide a detailed analysis of the channels activated by both STIM1 isoforms, revealing that STIM1L is more prone to open TRPC1, which might explain the larger SOCE elicited by this isoform.


Subject(s)
Calcium Signaling , Calcium/metabolism , Ion Channel Gating , Neoplasm Proteins/metabolism , Stromal Interaction Molecule 1/metabolism , TRPC Cation Channels/metabolism , Alternative Splicing/genetics , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , HEK293 Cells , Humans , Protein Binding
17.
Acta Physiol (Oxf) ; 228(4): e13417, 2020 04.
Article in English | MEDLINE | ID: mdl-31769918

ABSTRACT

AIM: It is widely accepted that the exocytosis of synaptic and secretory vesicles is triggered by Ca2+ entry through voltage-dependent Ca2+ channels. However, there is evidence of an alternative mode of exocytosis induced by membrane depolarization but lacking Ca2+ current and intracellular Ca2+ increase. In this work we investigated if such a mechanism contributes to secretory vesicle exocytosis in mouse chromaffin cells. METHODS: Exocytosis was evaluated by patch-clamp membrane capacitance measurements, carbon fibre amperometry and TIRF. Cytosolic Ca2+ was estimated using epifluorescence microscopy and fluo-8 (salt form). RESULTS: Cells stimulated by brief depolatizations in absence of extracellular Ca+2 show moderate but consistent exocytosis, even in presence of high cytosolic BAPTA concentration and pharmacological inhibition of Ca+2 release from intracellular stores. This exocytosis is tightly dependent on membrane potential, is inhibited by neurotoxin Bont-B (cleaves the v-SNARE synaptobrevin), is very fast (saturates with time constant <10 ms), it is followed by a fast endocytosis sensitive to the application of an anti-dynamin monoclonal antibody, and recovers after depletion in <5 s. Finally, this exocytosis was inhibited by: (i) ω-agatoxin IVA (blocks P/Q-type Ca2+ channel gating), (ii) in cells from knock-out P/Q-type Ca2+ channel mice, and (iii) transfection of free synprint peptide (interferes in P/Q channel-exocytic proteins association). CONCLUSION: We demonstrated that Ca2+ -independent and voltage-dependent exocytosis is present in chromaffin cells. This process is tightly coupled to membrane depolarization, and is able to support secretion during action potentials at low basal rates. P/Q-type Ca2+ channels can operate as voltage sensors of this process.


Subject(s)
Calcium Signaling/physiology , Chromaffin Cells/physiology , Exocytosis/physiology , Animals , Calcium/metabolism , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Female , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques/methods
18.
Methods Mol Biol ; 1942: 123-129, 2019.
Article in English | MEDLINE | ID: mdl-30900180

ABSTRACT

Calcium is a major regulator of neuronal activity and calcium signaling is critically important for normal neuronal function. Ca imaging is a well-established tool for studying neuronal function and ongoing spontaneous Ca2+ transients are a good indicator of neuronal maturity. There are various indicators available today, differing by their sensitivity, spectra, and loading method. Here we present a method for measurement of Ca2+ transients in neurons using two different Ca2+ indicators, Oregon Green BAPTA-1 and GCaMP6.


Subject(s)
Action Potentials , Calcium/metabolism , Cell Differentiation , Image Processing, Computer-Assisted/methods , Microscopy/methods , Neurons/metabolism , Calcium Signaling , Cells, Cultured , Chelating Agents/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Fluorescent Dyes/metabolism , Humans , Neurons/cytology , Organic Chemicals/metabolism
19.
Biochem Biophys Res Commun ; 507(1-4): 211-216, 2018 12 09.
Article in English | MEDLINE | ID: mdl-30415775

ABSTRACT

Ciliary beating frequency (CBF) was investigated in ciliated nasal epithelial cells (cMNECs) isolated from mice using video microscopy equipped with a high-speed camera. In cMNECs, a spontaneous CBF oscillation was observed. The CBF oscillation was abolished by BAPTA-AM but not by Ca2+-free solution. The addition of thapsigargin, which depletes Ca2+ from internal stores, also abolished CBF oscillation. Moreover, the intracellular Ca2+ concentration [Ca2+]i, spontaneously oscillated even with the Ca2+-free solution. Moreover, 2APB (an inhibitor of the IP3 receptor) abolished CBF oscillation in cMNECs. Overall, these findings suggest that the CBF oscillation in cMNECs is triggered by the release of Ca2+ from the IP3-sensitive internal stores. Moreover, IBMX, an inhibitor of phosphodiesterase, did not affect CBF oscillation in cMNECs, although it slightly increased CBF. These results suggest that CBF oscillations were induced by [Ca2+]i oscillation controlled via the release of Ca2+ from IP3-sensitive stores, rather than via cAMP accumulation. CBF oscillation possibly plays a crucial role in maintaining an efficient mucociliary clearance in the nasal epithelia.


Subject(s)
Calcium/metabolism , Cilia/metabolism , Intracellular Space/metabolism , Nasal Mucosa/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Boron Compounds/pharmacology , Cilia/drug effects , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Mice, Inbred C57BL , Nasal Mucosa/drug effects , Thapsigargin/pharmacology
20.
Cell Calcium ; 73: 82-87, 2018 07.
Article in English | MEDLINE | ID: mdl-29689523

ABSTRACT

The study of cellular Ca2+ signalling is indebted to Roger Tsien for the invention of fluorescent indicators that can be readily loaded into living cells and provide the means to measure cellular Ca2+ changes over long periods of time with sub-second resolution and microscopic precision. However, a recent study [1] reminds us that as useful as these tools are they need to be employed with caution as there can be off-target effects. This article summarises these recent findings within the wider context of confounding issues that can be encountered when using chemical and genetically-encoded Ca2+ indicators, and briefly discusses some approaches that may mitigate against misleading outcomes.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Cell Membrane/metabolism , Indicators and Reagents/metabolism , Animals , Calcium/analysis , Calcium Signaling/drug effects , Cell Membrane/chemistry , Cell Membrane/drug effects , Egtazic Acid/analogs & derivatives , Egtazic Acid/analysis , Egtazic Acid/metabolism , Egtazic Acid/pharmacology , Humans , Indicators and Reagents/analysis , Indicators and Reagents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL