1
|
De Pascali F, Inoue A, Benovic JL. Diverse pathways in GPCR-mediated activation of Ca 2+ mobilization in HEK293 cells. J Biol Chem 2024; 300:107882. [PMID: 39395798 DOI: 10.1016/j.jbc.2024.107882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024] Open
Abstract
G protein-coupled receptors transduce extracellular stimuli into intracellular signaling. Ca2+ is a well-known second messenger that can be induced by G protein-coupled receptor activation through the primary canonical pathways involving Gαq- and Gβγ-mediated activation of phospholipase C-β (PLCβ). While some Gs-coupled receptors are shown to trigger Ca2+ mobilization, underlying mechanisms remain elusive. Here, we evaluated whether Gs-coupled receptors including the β2-adrenergic receptor (β2AR) and the prostaglandin EP2 and EP4 receptors (EP2R and EP4R) that are endogenously expressed in human embryonic kidney 293 (HEK293) cells utilize common pathways for mediating Ca2+ mobilization. For the β2AR, we found an essential role for Gq in agonist-promoted Ca2+ mobilization while genetic or pharmacological inhibition of Gs or Gi had minimal effect. β-agonist-promoted Ca2+ mobilization was effectively blocked by the Gq-selective inhibitor YM-254890 and was not observed in ΔGαq/11 or ΔPLCβ cells. Bioluminescence resonance energy transfer analysis also suggests agonist-dependent association of the β2AR with Gq. For the EP2R, which couples to Gs, agonist treatment induced Ca2+ mobilization in a pertussis toxin-sensitive but YM-254890-insensitive manner. In contrast, EP4R, which couples to Gs and Gi, exhibited Ca2+ mobilization that was sensitive to both pertussis toxin and YM-254890. Interestingly, both EP2R and EP4R were largely unable to induce Ca2+ mobilization in ΔGαs or ΔPLCβ cells, supporting a strong dependency on Gs signaling in HEK293 cells. Taken together, we identify differences in the signaling pathways that are used to mediate Ca2+ mobilization in HEK293 cells where the β2AR primarily uses Gq, EP2R uses Gs and Gi, and EP4R uses Gs, Gi, and Gq.
Collapse
Affiliation(s)
- Francesco De Pascali
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
2
|
Mishra V, Adlakha N. Spatio temporal interdependent calcium and buffer dynamics regulating DAG in a hepatocyte cell due to obesity. J Bioenerg Biomembr 2023; 55:249-266. [PMID: 37460636 DOI: 10.1007/s10863-023-09973-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/23/2023] [Indexed: 09/12/2023]
Abstract
Calcium ions (Ca2+) serve as a crucial signaling mechanism in almost all cells. The buffers are proteins that bind free Ca2+ to reduce the cell's Ca2+ concentration. The most studies reported in the past on calcium signaling in various cells have considered the buffer concentration as constant in the cell. However, buffers also diffuse and their concentration varies dynamically in the cells. Almost no work has been reported on interdependent calcium and buffer dynamics in the cells. In the present study, a model is proposed for inter-dependent spatio-temporal dynamics of calcium and buffer by coupling reaction-diffusion equations of Ca2+ and buffer in a hepatocyte cell. Boundary and initial conditions are framed based on the physiological state of the cell. The effect of various parameters viz. inositol 1,4,5-triphosphate receptor (IP3R), diffusion coefficient, SERCA pump and ryanodine receptor (RyR) on spatio-temporal dynamics of calcium and buffer regulating diacylglycerol (DAG) in a normal and obese hepatocyte cell has been studied using finite element simulation. From the results, it is concluded that the dynamics of calcium and buffer impact each other significantly along the spatio-temporal dimensions, thereby affecting the regulation of all the processes including DAG in a hepatocyte cell. The proposed model is more realistic than the existing ones, as the interdependent system dynamics of calcium and buffer have different regulatory impacts as compared to the individual and independent dynamics of these signaling processes in a hepatocyte cell.
Collapse
Affiliation(s)
- Vedika Mishra
- Department of Mathematics, SVNIT, Gujarat, 395007, Surat, India.
| | - Neeru Adlakha
- Department of Mathematics, SVNIT, Gujarat, 395007, Surat, India
| |
Collapse
|
3
|
Wang R, Wu Y, Zhu Y, Yao S, Zhu Y. ANKRD22 is a novel therapeutic target for gastric mucosal injury. Pharmacotherapy 2022; 147:112649. [DOI: 10.1016/j.biopha.2022.112649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/28/2022]
|
4
|
Umemoto S, Furusawa T, Unuma H, Tajika M, Sekino T. In vivo bioresorbability and bone formation ability of sintered highly pure calcium carbonate granules. Dent Mater J 2021; 40:1202-1207. [PMID: 34121021 DOI: 10.4012/dmj.2020-254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Calcium carbonate-based bone substitutes derived from natural coral exoskeleton (aragonite) are resorbed and remodeled faster than calcium phosphate-based substitutes. However, coral species with structures appropriate for use as bone substitutes are very limited. Therefore, it is important to evaluate potential of artificial calcium carbonate ceramics as a bone substitute. In this study, calcium carbonate granules with various porosities and pore sizes were prepared by sintering a highly pure (>99.98%) calcium carbonate powder (calcite), and their resorption properties and bone formation abilities were examined in vivo for the first time. The sintered calcium carbonate was resorbed faster than β-tricalcium phosphate, which has a similar structure. However, sintered calcium carbonate did not promote new bone formation during long-term implantation. Furthermore, both resorption and new bone formation were affected by the pore structure. The optimal structures of the artificially sintered calcium carbonate bone substitute were also discussed.
Collapse
Affiliation(s)
- Shota Umemoto
- Shiraishi Central Laboratories Co., Ltd.,The Institute of Scientific and Industrial Research, Osaka University
| | - Toshitake Furusawa
- Tohoku Oral Implant Association.,Graduate School of Science and Engineering, Yamagata University
| | - Hidero Unuma
- Tohoku Oral Implant Association.,Graduate School of Science and Engineering, Yamagata University
| | | | - Tohru Sekino
- The Institute of Scientific and Industrial Research, Osaka University
| |
Collapse
|
5
|
Li C, Qian T, He R, Wan C, Liu Y, Yu H. Endoplasmic Reticulum-Plasma Membrane Contact Sites: Regulators, Mechanisms, and Physiological Functions. Front Cell Dev Biol 2021; 9:627700. [PMID: 33614657 PMCID: PMC7889955 DOI: 10.3389/fcell.2021.627700] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum (ER) forms direct membrane contact sites with the plasma membrane (PM) in eukaryotic cells. These ER-PM contact sites play essential roles in lipid homeostasis, ion dynamics, and cell signaling, which are carried out by protein-protein or protein-lipid interactions. Distinct tethering factors dynamically control the architecture of ER-PM junctions in response to intracellular signals or external stimuli. The physiological roles of ER-PM contact sites are dependent on a variety of regulators that individually or cooperatively perform functions in diverse cellular processes. This review focuses on proteins functioning at ER-PM contact sites and highlights the recent progress in their mechanisms and physiological roles.
Collapse
Affiliation(s)
- Chenlu Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tiantian Qian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
6
|
Marumo M, Ekawa K, Wakabayashi I. Resveratrol inhibits Ca 2+ signals and aggregation of platelets. Environ Health Prev Med 2020; 25:70. [PMID: 33160329 PMCID: PMC7648989 DOI: 10.1186/s12199-020-00905-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/25/2020] [Indexed: 11/10/2022] Open
Abstract
Background Resveratrol has been shown to inhibit platelet aggregation. However, the mechanism for this action of resveratrol remains to be clarified. The purpose of this study was to elucidate the Ca2+-related mechanism for the inhibitory action of resveratrol on platelet aggregation. Methods Ca2+ entry and subsequent aggregation of human platelets induced by different stimulants including thrombin, thapsigargin, and 1-oleoyl-2-acetylglycerol (OAG) were measured by the fluorescence method and light transmittance method, respectively. Each stimulant was added to a nominally Ca2+-free medium containing platelets, and then CaCl2 was added to the medium to induce Ca2+ influx into platelets. Results Thapsigargin-induced Ca2+ entry into platelets and subsequent platelet aggregation were significantly inhibited in the presence of resveratrol at 6.25 μM or higher concentrations, while OAG-induced Ca2+ entry and subsequent platelet aggregation were not affected by resveratrol at concentrations up to 50 μM. In the nominally Ca2+-free medium, thrombin induced a small transient increase in intracellular Ca2+ concentrations, which was attenuated in the presence of resveratrol at 12.5 μM or higher concentrations. Thrombin-induced Ca2+ entry into platelets and subsequent platelet aggregation were significantly inhibited in the presence of resveratrol at 12.5 μM or higher concentrations. Conclusions The results suggest that resveratrol inhibits thrombin-induced platelet aggregation through decreasing Ca2+ release from its stores and inhibiting store-operated Ca2+ influx into platelets.
Collapse
Affiliation(s)
- Mikio Marumo
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kazumi Ekawa
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo, 663-8501, Japan
| | - Ichiro Wakabayashi
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
7
|
Chowdhury SM, Xie S, Fang J, Lee SK, Sitharaman B. Nanoparticle-Facilitated Membrane Depolarization-Induced Receptor Activation: Implications on Cellular Uptake and Drug Delivery. ACS Biomater Sci Eng 2016; 2:2153-2161. [PMID: 33465891 DOI: 10.1021/acsbiomaterials.6b00338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cell-specific uptake of drug delivery systems (DDSs) are crucial to achieve optimal efficacy of many drugs. Widely employed strategies to facilitate targeted intracellular drug delivery involves attachment of targeting ligands (peptides or antibodies) to DDSs. Target receptors mutations can limit the effectiveness of this approach. Herein, we demonstrate, through in vitro inhibitory and drug delivery studies, that graphene nanoribbons (GNRs), water dispersed with the amphiphilic polymer called PEG-DSPE ((1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N [amino (polyethylene glycol)]) (induce membrane depolarization-mediated epidermal growth factor receptor (EGFR) activation. This phenomenon is ligand-independent and EGFR activation occurs via influx of Ca2+ ions from the extracellular space. We further provide evidence, through in vivo studies, that this mechanism could be exploited to facilitate efficacious drug delivery into tumors that overexpress EGFR. The results suggest that transient membrane depolarization-facilitated cell receptor activation can be employed as an alternate strategy for enhanced intracellular drug delivery.
Collapse
Affiliation(s)
- Sayan Mullick Chowdhury
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Shawn Xie
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Justin Fang
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Stephen K Lee
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Balaji Sitharaman
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
8
|
Cebotaru L, Cebotaru V, Wang H, Arend LJ, Guggino WB. STIM1fl/fl Ksp-Cre Mouse has Impaired Renal Water Balance. Cell Physiol Biochem 2016; 39:172-82. [PMID: 27336410 DOI: 10.1159/000445614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/AIM STIM1 is as an essential component in store operated Ca2+ entry. However give the paucity of information on the role of STIM1 in kidney, the aim was to study the function of STIM1 in the medulla of the kidney. METHODS we crossed a Ksp-cre mouse with another mouse containing two loxP sites flanking Exon 6 of STIM1. The Ksp-cre mouse is based upon the Ksp-cadherin gene promoter which expresses cre recombinase in developing nephrons, collecting ducts (SD) and thick ascending limbs (TAL) of the loop of Henle. RESULTS The offspring of these mice are viable without gross morphological changes, however, we noticed that the STIM1 Ksp-cre knockout mice produced more urine compared to control. To examine this more carefully, we fed mice low (LP) and high protein (HP) diets respectively. When mice were fed HP diet STIM1 ko mice had significantly increased urinary volume and lower specific gravity compared to wt mice. In STIM1 ko mice fed HP diet urine creatinine and urea were significantly lower compared to wt mice fed HP diet, however the fractional excretion was the same. CONCLUSION These data support the idea that STIM1 ko mice have impaired urinary concentrating ability when challenged with HP diet is most likely caused by impaired Ca2+-dependent signal transduction through the vasopressin receptor cascade.
Collapse
Affiliation(s)
- Liudmila Cebotaru
- Division of Gastroenterology, Department of Medicine, Division of Nephrology, University of Maryland, Baltimore, USA
| | | | | | | | | |
Collapse
|
9
|
Chen CY, Liaw CC, Chen YH, Chang WY, Chung PJ, Hwang TL. A novel immunomodulatory effect of ugonin U in human neutrophils via stimulation of phospholipase C. Free Radic Biol Med 2014; 72:222-31. [PMID: 24747490 DOI: 10.1016/j.freeradbiomed.2014.04.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 04/10/2014] [Accepted: 04/14/2014] [Indexed: 11/19/2022]
Abstract
Neutrophils have a crucial role in the immune system and are the first line of defense against pathogenic invaders. Neutrophil activation is required for their defensive function and can be induced by diverse stimuli, through either binding to cell surface receptors or direct intracellular target molecule stimulation. In this study, we found that 4″a,5″,6″,7″,8″,8″a-hexahydro-5,3',4'-trihydroxy-5″,5″,8″a-trimethyl-4H-chromeno [2″,3″:7,6]flavone (ugonin U), a flavonoid isolated from Helminthostachys zeylanica (L) Hook, significantly induced superoxide production and release in a time- and concentration-dependent manner. A series of experiments was performed to dissect the mechanism of ugonin U-induced respiratory burst in human neutrophils. Our results demonstrated that ugonin U induced a slow increase in intracellular Ca(2+), which was necessary for ugonin U-stimulated superoxide release. Use of a formyl peptide receptor (FPR) blocker, G protein inhibitor, and protein tyrosine kinase (PTK) inhibitor proved that FPR, G proteins, and PTKs were not associated with ugonin U-induced respiratory burst. Additionally, immunoblotting results revealed that ugonin U did not affect the phosphorylation of mitogen-activated protein kinases and protein tyrosine. Nevertheless, a phospholipase C (PLC) inhibitor and an inositol triphosphate (IP3) receptor antagonist considerably suppressed ugonin U-stimulated Ca(2+) mobilization and subsequent superoxide release. Ugonin U also induced an increase in intracellular IP3 formation, which could be blocked using a PLC inhibitor. In conclusion, our study reveals that ugonin U represents the first identified natural flavonoid compound to directly stimulate PLC. Moreover, ugonin U induces respiratory burst via the PLC/IP3/Ca(2+) pathway in human neutrophils.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Graduate Institute of Natural Products, School of Traditional Medicine, College of Medicine, Chang Gung University, Kweishan 333, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kweishan 333, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chih-Chuang Liaw
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yi-Hsuan Chen
- Graduate Institute of Natural Products, School of Traditional Medicine, College of Medicine, Chang Gung University, Kweishan 333, Taoyuan, Taiwan
| | - Wen-Yi Chang
- Graduate Institute of Natural Products, School of Traditional Medicine, College of Medicine, Chang Gung University, Kweishan 333, Taoyuan, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan 333, Taoyuan, Taiwan
| | - Pei-Jen Chung
- Graduate Institute of Natural Products, School of Traditional Medicine, College of Medicine, Chang Gung University, Kweishan 333, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, School of Traditional Medicine, College of Medicine, Chang Gung University, Kweishan 333, Taoyuan, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan 333, Taoyuan, Taiwan.
| |
Collapse
|
10
|
Derler I, Fritsch R, Schindl R, Romanin C. CRAC inhibitors: identification and potential. Expert Opin Drug Discov 2013; 3:787-800. [PMID: 23496221 DOI: 10.1517/17460441.3.7.787] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Ca(2+) release-activated Ca(2+) (CRAC) channels, a subfamily of store-operated channels, play an essential role in various diseases such as immune disorders and allergic responses. OBJECTIVE The successful treatment of these diseases requires the identification of specific inhibitors. So far, a variety of chemical compounds blocking CRAC have been identified; however, they have all turned out to be less specific. Recently two proteins, STIM1 and ORAI1, have been identified as the essential components that fully reconstitute CRAC currents with a similar biophysical fingerprint. METHOD These two proteins and their activation process represent direct targets for the application of specific CRAC inhibitors. RESULTS/CONCLUSION For drug development, fluorescence microscopy adaptable for high-throughput screening will provide a powerful assay to mechanistically identify potential CRAC inhibitors that act on various stages within the STIM1/ORAI1 activation pathway visualized by fluorescent-tagged proteins.
Collapse
Affiliation(s)
- Isabella Derler
- University of Linz, Institute of Biophysics, A-4040 Linz, Austria +43 732 2468 9272 ; +43 732 2468 9280 ; ;
| | | | | | | |
Collapse
|
11
|
Li X, Li PCH. Strategies for the real-time detection of Ca2+ channel events of single cells: recent advances and new possibilities. Expert Rev Clin Pharmacol 2012; 3:267-80. [PMID: 22111609 DOI: 10.1586/ecp.10.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ca(2+) ion channels play key roles in cell physiology and they are important drug targets. The Ca(2+) channel events are mainly measurable by fluorescent and patch clamp methods. This review summarizes the recent advances of these techniques for the detection of Ca(2+) channel events and the prospect of their new directions in the near future. Conventional bulk fluorescent methods are amenable to high-throughput applications, but they are not real-time single-cell measurements, which provide kinetic data on individual cells and offer unparalleled sensitive data for rare cells. Recent advances on real-time single-cell fluorescent measurements are conducted on microfluidic chips with scalable cell-retention sites, integrated with electrical stimulation and fluorescent measuring features. Patch clamp techniques are real-time measurements conducted on single cells, but the measurements are of low throughput. Recent advances are conducted on microfluidic patch clamp chips for high-throughput applications. Future real-time single-cell Ca(2+) channel event measurements will be conducted in a multiparametric manner in an integrated and automated microfluidic chip.
Collapse
Affiliation(s)
- XiuJun Li
- University of California at Berkeley, CA 94720, USA
| | | |
Collapse
|
12
|
Structure, regulation and biophysics of I(CRAC), STIM/Orai1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:383-410. [PMID: 22453951 DOI: 10.1007/978-94-007-2888-2_16] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ca(2+) release activated Ca(2+) (CRAC) channels mediate robust Ca(2+) influx when the endoplasmic reticulum Ca(2+) stores are depleted. This essential process for T-cell activation as well as degranulation of mast cells involves the Ca(2+) sensor STIM1, located in the endoplasmic reticulum and the Ca(2+) selective Orai1 channel in the plasma membrane. Our review describes the CRAC signaling pathway, the activation of which is initiated by a drop in the endoplasmic Ca(2+) level sensed by STIM1. This in term induces multimerisation and puncta-formation of STIM1 proteins is followed by their coupling to and activation of Orai channels. Consequently Ca(2+) entry is triggered through the Orai pore into the cytosol with subsequent closure of the channel by Ca(2+)-dependent inactivation. We will portray a mechanistic view of the events coupling STIM1 to Orai activation based on their structure and biophysics.
Collapse
|
13
|
|
14
|
Diacylglycerol signaling underlies astrocytic ATP release. Neural Plast 2011; 2011:537659. [PMID: 21826278 PMCID: PMC3151491 DOI: 10.1155/2011/537659] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 05/05/2011] [Indexed: 01/31/2023] Open
Abstract
Astrocytes have the ability to modulate neuronal excitability and synaptic transmission by the release of gliotransmitters. The importance of ATP released downstream of the activation of Gq-coupled receptors has been well established, but the mechanisms by which this release is regulated are unclear. The current work reveals that the elevation of diacylglycerol (DAG) in astrocytes induces vesicular ATP release. Unexpectedly, DAG-induced ATP release was found to be independent of PKC activation, but dependent upon activation of a C1 domain-containing protein. Astrocytes express the C1 domain-containing protein Munc13-1, which has been implicated in neuronal transmitter release, and RNAi-targeted downregulation of Munc13-1 inhibits astrocytic ATP release. These studies demonstrate that elevations of DAG induce the exocytotic release of ATP in astrocytes, likely via a Munc13-1-dependent mechanism.
Collapse
|
15
|
Mumtaz S, Burdyga G, Borisova L, Wray S, Burdyga T. The mechanism of agonist induced Ca2+ signalling in intact endothelial cells studied confocally in in situ arteries. Cell Calcium 2010; 49:66-77. [PMID: 21176847 PMCID: PMC3098389 DOI: 10.1016/j.ceca.2010.11.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 11/24/2010] [Accepted: 11/25/2010] [Indexed: 11/25/2022]
Abstract
In endothelial cells there remain uncertainties in the details of how Ca2+ signals are generated and maintained, especially in intact preparations. In particular the role of the sarco-endoplasmic reticulum Ca2+-ATPase (SERCA), in contributing to the components of agonist-induced signals is unclear. The aim of this work was to increase understanding of the detailed mechanism of Ca2+ signalling in endothelial cells using real time confocal imaging of Fluo-4 loaded intact rat tail arteries in response to muscarinic stimulation. In particular we have focused on the role of SERCA, and its interplay with capacitative Ca2+ entry (CCE) and ER Ca2+ release and uptake. We have determined its contribution to the Ca2+ signal and how it varies with different physiological stimuli, including single and repeated carbachol applications and brief and prolonged exposures. In agreement with previous work, carbachol stimulated a rise in intracellular Ca2+ in the endothelial cells, consisting of a rapid initial phase, then a plateau upon which oscillations of Ca2+ were superimposed, followed by a decline to basal Ca2+ levels upon carbachol removal. Our data support the following conclusions: (i) the size (amplitude and duration) of the Ca2+ spike and early oscillations are limited by SERCA activity, thus both are increased if SERCA is inhibited. (ii) SERCA activity is such that brief applications of carbachol do not trigger CCE, presumably because the fall in luminal Ca2+ is not sufficient to trigger it. However, longer applications sufficient to deplete the ER or even partial SERCA inhibition stimulate CCE. (iii) Ca2+ entry occurs via STIM-mediated CCE and SERCA contributes to the cessation of CCE. In conclusion our data show how SERCA function is crucial to shaping endothelial cell Ca signals and its dynamic interplay with both CCE and ER Ca releases.
Collapse
Affiliation(s)
- S Mumtaz
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, UK
| | | | | | | | | |
Collapse
|
16
|
Cerella C, Cordisco S, Albertini MC, Accorsi A, Diederich M, Ghibelli L. Magnetic fields promote a pro-survival non-capacitative Ca2+ entry via phospholipase C signaling. Int J Biochem Cell Biol 2010; 43:393-400. [PMID: 21095240 DOI: 10.1016/j.biocel.2010.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 11/04/2010] [Accepted: 11/15/2010] [Indexed: 11/28/2022]
Abstract
The ability of magnetic fields (MFs) to promote/increase Ca(2+) influx into cells is widely recognized, but the underlying mechanisms remain obscure. Here we analyze how static MFs of 6 mT modulates thapsigargin-induced Ca(2+) movements in non-excitable U937 monocytes, and how this relates to the anti-apoptotic effect of MFs. Magnetic fields do not affect thapsigargin-induced Ca(2+) mobilization from endoplasmic reticulum, but significantly increase the resulting Ca(2+) influx; this increase requires intracellular signal transduction actors including G protein, phospholipase C, diacylglycerol lipase and nitric oxide synthase, and behaves as a non-capacitative Ca(2+) entry (NCCE), a type of influx with an inherent signaling function, rather than a capacitative Ca(2+) entry (CCE). All treatments abrogating the extra Ca(2+) influx also abrogate the anti-apoptotic effect of MFs, demonstrating that MF-induced NCCE elicits an anti-apoptotic survival pathway.
Collapse
Affiliation(s)
- Claudia Cerella
- Dipartimento di Biologia, Universita' degli Studi di Roma Tor Vergata, Via della Ricerca Scientifica snc, 00133 Roma, Italy
| | | | | | | | | | | |
Collapse
|
17
|
Liu W, Tang F, Chen J. Designing dynamical output feedback controllers for store-operated Ca²+ entry. Math Biosci 2010; 228:110-8. [PMID: 20816868 DOI: 10.1016/j.mbs.2010.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2009] [Revised: 06/21/2010] [Accepted: 08/27/2010] [Indexed: 12/26/2022]
Abstract
Store-operated calcium entry (SOCE) has been proposed as the main process controlling Ca²+ entry in non-excitable cells. Although recent breakthroughs in experimental studies of SOCE have been made, its mathematical modeling has not been developed. In the present work, SOCE is viewed as a feedback control system subject to an extracellular agonist disturbance and an extracellular calcium input. We then design a dynamic output feedback controller to reject the disturbance and track Ca²+ resting levels in the cytosol and the endoplasmic reticulum (ER). The constructed feedback control system is validated by published experimental data and its global asymptotic stability is proved by using the LaSalle's invariance principle. We then simulate the dynamic responses of STIM1 and Orai1, two major components in the operation of the store-operated channels, to the depletion of Ca²+ in the ER with thapsigargin, which show that: (1) Upon the depletion of Ca²+ in the ER, the concentrations of activated STIM1 and STIM1-Orai1 cluster are elevated gradually, indicating that STIM1 is accumulating in the ER-PM junctions and that the cytosolic portion of the active STIM1 is binding to Orai1 and driving the opening of CRAC channels for Ca²+ entry; (2) after the extracellular Ca²+ addition, the concentrations of both STIM1 and STIM1-Orai1 cluster decrease but still much higher than the original levels. We also simulate the system responses to the agonist disturbance, which show that, when a sequence of periodic agonist pulses is applied, the system returns to its equilibrium after each pulse. This indicates that the designed feedback controller can reject the disturbance and track the equilibrium.
Collapse
Affiliation(s)
- Weijiu Liu
- Department of Mathematics, University of Central Arkansas, 201 Donaghey Avenue, Conway, AR 72035, USA.
| | | | | |
Collapse
|
18
|
Arvizo RR, Miranda OR, Thompson MA, Pabelick CM, Bhattacharya R, Robertson JD, Rotello VM, Prakash Y, Mukherjee P. Effect of nanoparticle surface charge at the plasma membrane and beyond. NANO LETTERS 2010; 10:2543-8. [PMID: 20533851 PMCID: PMC2925219 DOI: 10.1021/nl101140t] [Citation(s) in RCA: 410] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Herein, we demonstrate that the surface charge of gold nanoparticles (AuNPs) plays a critical role in modulating membrane potential of different malignant and nonmalignant cell types and subsequent downstream intracellular events. The findings presented here describe a novel mechanism for cell-nanoparticle interactions and AuNP uptake: modulation of membrane potential and its effect on intracellular events. These studies will help understand the biology of cell-nanoparticle interactions and facilitate the engineering of nanoparticles for specific intracellular targets.
Collapse
Affiliation(s)
- Rochelle R. Arvizo
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Oscar R. Miranda
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003
| | - Michael A. Thompson
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Christina M. Pabelick
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Resham Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - J. David Robertson
- Department of Chemistry and University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211
| | | | - Y.S. Prakash
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Priyabrata Mukherjee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN 55905
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, MN 55905
| |
Collapse
|
19
|
Komarova Y, Malik AB. Regulation of endothelial permeability via paracellular and transcellular transport pathways. Annu Rev Physiol 2010; 72:463-93. [PMID: 20148685 DOI: 10.1146/annurev-physiol-021909-135833] [Citation(s) in RCA: 480] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The endothelium functions as a semipermeable barrier regulating tissue fluid homeostasis and transmigration of leukocytes and providing essential nutrients across the vessel wall. Transport of plasma proteins and solutes across the endothelium involves two different routes: one transcellular, via caveolae-mediated vesicular transport, and the other paracellular, through interendothelial junctions. The permeability of the endothelial barrier is an exquisitely regulated process in the resting state and in response to extracellular stimuli and mediators. The focus of this review is to provide a comprehensive overview of molecular and signaling mechanisms regulating endothelial barrier permeability with emphasis on the cross-talk between paracellular and transcellular transport pathways.
Collapse
Affiliation(s)
- Yulia Komarova
- Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | |
Collapse
|
20
|
Activation of the inositol (1,4,5)-triphosphate calcium gate receptor is required for HIV-1 Gag release. J Virol 2010; 84:6438-51. [PMID: 20427533 DOI: 10.1128/jvi.01588-09] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The structural precursor polyprotein, Gag, encoded by all retroviruses, including the human immunodeficiency virus type 1 (HIV-1), is necessary and sufficient for the assembly and release of particles that morphologically resemble immature virus particles. Previous studies have shown that the addition of Ca(2+) to cells expressing Gag enhances virus particle production. However, no specific cellular factor has been implicated as mediator of Ca(2+) provision. The inositol (1,4,5)-triphosphate receptor (IP3R) gates intracellular Ca(2+) stores. Following activation by binding of its ligand, IP3, it releases Ca(2+) from the stores. We demonstrate here that IP3R function is required for efficient release of HIV-1 virus particles. Depletion of IP3R by small interfering RNA, sequestration of its activating ligand by expression of a mutated fragment of IP3R that binds IP3 with very high affinity, or blocking formation of the ligand by inhibiting phospholipase C-mediated hydrolysis of the precursor, phosphatidylinositol-4,5-biphosphate, inhibited Gag particle release. These disruptions, as well as interference with ligand-receptor interaction using antibody targeted to the ligand-binding site on IP3R, blocked plasma membrane accumulation of Gag. These findings identify IP3R as a new determinant in HIV-1 trafficking during Gag assembly and introduce IP3R-regulated Ca(2+) signaling as a potential novel cofactor in viral particle release.
Collapse
|
21
|
Martini A, Bruno R, Mazzulla S, Nocita A, Martino G. Angiotensin II regulates endothelial cell migration through calcium influx via T-type calcium channel in human umbilical vein endothelial cells. Acta Physiol (Oxf) 2010; 198:449-55. [PMID: 20028346 DOI: 10.1111/j.1748-1716.2009.02070.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AIM The T-type calcium channel is expressed in vascular endothelial cells, but its role in endothelial cell function is yet to be elucidated. We analysed the endothelial functional role of T-type calcium channel-dependent calcium under angiotensin II (Ang II) stimulation. METHODS Human umbilical vein endothelial cells were co-incubated with hormone at 10(-7) m and either Efonidipine 10(-5) m or Verapamil 10(-5) m or Mibefradil 10(-5) m or Wortmannin 10(-6) m. The contribution of Ang II receptors was evaluated using PD123319 10(-7) m and ZD 7155 10(-7) m. The calcium ion concentration was observed using Fluo-3 acetossimetil ester. The cells were observed after 3, 6, 9 and 12 h. RESULTS The microfluorescence method points out that Ang II induces intracellular calcium modulation in time by distinct mechanisms. AT2 receptor blockade is necessary to observe significant increase in [Ca(2+)](i) levels. Pre-treatment with Mibefradil abolishes Ang II -induced cell migration. CONCLUSIONS Our data show that Ang II, via AT1 receptor, modulates calcium concentration involving T-type calcium channel and L-type calcium channel but only the calcium influx via T-type calcium channels regulates endothelial cell migration which is essential for angiogenesis.
Collapse
|
22
|
Chen JB, Tao R, Sun HY, Tse HF, Lau CP, Li GR. Multiple Ca2+ signaling pathways regulate intracellular Ca2+ activity in human cardiac fibroblasts. J Cell Physiol 2010; 223:68-75. [PMID: 20039269 DOI: 10.1002/jcp.22010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Ca(2+) signaling pathways are well studied in cardiac myocytes, but not in cardiac fibroblasts. The aim of the present study is to characterize Ca(2+) signaling pathways in cultured human cardiac fibroblasts using confocal scanning microscope and RT-PCR techniques. It was found that spontaneous intracellular Ca(2+) (Ca(i) (2+)) oscillations were present in about 29% of human cardiac fibroblasts, and the number of cells with Ca(i) (2+) oscillations was increased to 57.3% by application of 3% fetal bovine serum. Ca(i) (2+) oscillations were dependent on Ca(2+) entry. Ca(i) (2+) oscillations were abolished by the store-operated Ca(2+) (SOC) entry channel blocker La(3+), the phospholipase C inhibitor U-73122, and the inositol trisphosphate receptors (IP3Rs) inhibitor 2-aminoethoxydiphenyl borate, but not by ryanodine. The IP3R agonist thimerosal enhanced Ca(i) (2+) oscillations. Inhibition of plasma membrane Ca(2+) pump (PMCA) and Na(+)-Ca(2+) exchanger (NCX) also suppressed Ca(i) (2+) oscillations. In addition, the frequency of Ca(i) (2+) oscillations was reduced by nifedipine, and increased by Bay K8644 in cells with spontaneous Ca(2+) oscillations. RT-PCR revealed that mRNAs for IP3R1-3, SERCA1-3, Ca(V)1.2, NCX3, PMCA1,3,4, TRPC1,3,4,6, STIM1, and Orai1-3, were readily detectable, but not RyRs. Our results demonstrate for the first time that spontaneous Ca(i) (2+) oscillations are present in cultured human cardiac fibroblasts and are regulated by multiple Ca(2+) pathways, which are not identical to those of the well-studied contractile cardiomyocytes. This study provides a base for future investigations into how Ca(2+) signals regulate biological activity in human cardiac fibroblasts and cardiac remodeling under pathological conditions.
Collapse
Affiliation(s)
- Jing-Bo Chen
- Li Ka Shing Faculty of Medicine, Department of Medicine, Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
23
|
Hu R, He ML, Hu H, Yuan BX, Zang WJ, Lau CP, Tse HF, Li GR. Characterization of calcium signaling pathways in human preadipocytes. J Cell Physiol 2009; 220:765-70. [DOI: 10.1002/jcp.21823] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
24
|
Activation of phosphatidylinositol-linked D1-like receptor modulates FGF-2 expression in astrocytes via IP3-dependent Ca2+ signaling. J Neurosci 2009; 29:7766-75. [PMID: 19535588 DOI: 10.1523/jneurosci.0389-09.2009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fibroblast growth factor-2 (FGF-2) is predominantly synthesized and secreted by astrocytes in adult brain. Our previous study showed that activation of classical dopamine receptor D(1) or D(2) elicits FGF-2 biosynthesis and secretion in astrocytes. Here, we report that astrocytic FGF-2 expression is also regulated by phosphatidylinositol (PI)-linked D(1)-like receptor. SKF83959, a selective PI-linked D(1)-like receptor agonist, upregulates the levels of FGF-2 protein in striatal astrocyte cultures in classical dopamine D(1) and D(2) receptor-independent manner. The conditional medium derived from SKF83959-activated astrocytes promoted the number of TH(+) neurons in vitro. Treatment of astrocytes with SKF83959 increased intracellular calcium in two phases. Inhibition of intracellular calcium oscillation by inositol 1,4,5-triphosphate (IP3) inhibitors blocked the SKF83959-induced increase in FGF-2 expression. Moreover, intraperitoneal administration of SKF83959 reversed l-methyl-4-phenyl-l,2,3,6-tetrahydropypridine (MPTP)-induced reduction in FGF-2 expression in both the striatum and ventral midbrain and resulted in marked protection of dopaminergic neurons from MPTP-induced neurotoxicity. These results indicate that IP3/Ca(2+)/calmodulin-dependent protein kinase is an uncharted intracellular signaling pathway that is crucial for the regulation of FGF-2 synthesis in astrocytes. PI-linked D(1)-like receptor plays an important role in the regulation of astrocytic FGF-2 expression and neuroprotection which may provide a potential target for the drug discovery in Parkinson's disease.
Collapse
|
25
|
Gomez-Pinilla PJ, Camello PJ, Pozo MJ. Pancreatic calcium signaling: role in health and disease. Pancreatology 2009; 9:329-33. [PMID: 19451741 DOI: 10.1159/000213412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In order to control cell functions, extracellular agents, such as hormones or neurotransmitters among others, generate a diversity of calcium (Ca(2+)) signals in target cells. Here, we review the components involved in Ca(2+) handling and effectors, both members of the known calcium signaling pathways. In the pancreas, Ca(2+) signal appears as local increases, global elevations or Ca(2+) oscillations. Ca(2+) plays a key role in the pancreatic cells, regulating secretion in exocrine cells, a widely used model for studying the coupling between Ca(2+) signaling and secretion, and the release of insulin, glucagon and somatostatin in the exocrine pancreas. Interestingly, Ca(2+) deregulations have been related to pancreatitis and aging of the pancreas, and treatment with melatonin has shown beneficial effects suggesting that melatonin could be an adequate therapeutic approach.
Collapse
Affiliation(s)
- Pedro J Gomez-Pinilla
- Department of Physiology, Nursing School, University of Extremadura, Cáceres, Spain.
| | | | | |
Collapse
|
26
|
Derler I, Fahrner M, Carugo O, Muik M, Bergsmann J, Schindl R, Frischauf I, Eshaghi S, Romanin C. Increased hydrophobicity at the N terminus/membrane interface impairs gating of the severe combined immunodeficiency-related ORAI1 mutant. J Biol Chem 2009; 284:15903-15. [PMID: 19366689 DOI: 10.1074/jbc.m808312200] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Patients with severe combined immune deficiency (SCID) suffer from defective T-cell Ca2+ signaling. A loss of Ca2+ entry has been linked at the molecular level to single missense mutation R91W in the store-operated Ca2+ channel ORAI1. However, the mechanistic impact of this mutation on ORAI1 function remains unclear. Confocal Förster resonance energy transfer microscopy revealed that dynamic store-operated coupling of STIM1 to ORAI1 R91W was largely sustained similar to wild-type ORAI1. Characterization of various point mutants at position 91 by whole cell patch clamp recordings displayed that neutral or even negatively charged amino acids did not abolish ORAI1 function. However, substitution by hydrophobic leucine, valine, or phenylalanine resulted in non-functional ORAI1 channels, despite preserved STIM1 coupling. Besides conformational constraints at the N terminus/membrane interface predicted for the hydrophobic mutants, additional key factor(s) were suggested to determine ORAI1 functionality. Calculation of the probability for the 1st transmembrane domain and its hydrophobicity revealed a substantial increase for all hydrophobic substitutions that lead to non-functional ORAI1 R91X mutants in contrast to those with hydrophilic residues. Hence, increased hydrophobicity might lead to disrupted permeation/gating, as an ORAI1 channel with increased pore size and R91W mutation failed to recover activity. In conclusion, the increase in hydrophobicity at the N terminus/membrane interface represents the major cause for yielding non-functional ORAI1 channels.
Collapse
Affiliation(s)
- Isabella Derler
- Institute for Biophysics, University of Linz, Altenbergerstrasse 69, 4040 Linz, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lebiedzinska M, Szabadkai G, Jones AWE, Duszynski J, Wieckowski MR. Interactions between the endoplasmic reticulum, mitochondria, plasma membrane and other subcellular organelles. Int J Biochem Cell Biol 2009; 41:1805-16. [PMID: 19703651 DOI: 10.1016/j.biocel.2009.02.017] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Revised: 02/22/2009] [Accepted: 02/23/2009] [Indexed: 02/02/2023]
Abstract
Several recent works show structurally and functionally dynamic contacts between mitochondria, the plasma membrane, the endoplasmic reticulum, and other subcellular organelles. Many cellular processes require proper cooperation between the plasma membrane, the nucleus and subcellular vesicular/tubular networks such as mitochondria and the endoplasmic reticulum. It has been suggested that such contacts are crucial for the synthesis and intracellular transport of phospholipids as well as for intracellular Ca(2+) homeostasis, controlling fundamental processes like motility and contraction, secretion, cell growth, proliferation and apoptosis. Close contacts between smooth sub-domains of the endoplasmic reticulum and mitochondria have been shown to be required also for maintaining mitochondrial structure. The overall distance between the associating organelle membranes as quantified by electron microscopy is small enough to allow contact formation by proteins present on their surfaces, allowing and regulating their interactions. In this review we give a historical overview of studies on organelle interactions, and summarize the present knowledge and hypotheses concerning their regulation and (patho)physiological consequences.
Collapse
|
28
|
Barajas M, Andrade A, Hernandez-Hernandez O, Felix R, Arias-Montaño JA. Histamine-induced Ca2+ entry in human astrocytoma U373 MG cells: evidence for involvement of store-operated channels. J Neurosci Res 2009; 86:3456-68. [PMID: 18627030 DOI: 10.1002/jnr.21784] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Glial and glia-derived cells express a variety of receptors for neurotransmitters and hormones, the majority of which evoke both Ca(2+) release from intracellular stores and Ca(2+) entry across the plasma membrane. We investigated the links between histamine H(1) receptor activation, Ca(2+) release from intracellular stores and Ca(2+) influx in human astrocytoma U373 MG cells. Histamine, through a H(1) receptor-mediated effect, evoked an increase in cytoplasmic free calcium concentration ([Ca(2+)](i)) that occurred in two phases: an initial, transient, increase owing to Ca(2+) mobilization from intracellular pools, and a second, sustained increase dependent on both Ca(2+) influx and continuous receptor occupancy. The characteristics of histamine-induced increases in [Ca(2+)](i) were similar to the capacitative entry evoked by emptying of the Ca(2+) stores with thapsigargine, and different from that observed when Ca(2+) influx was activated with OAG (1-oleoyl-2-acetyl-sn-glycerol), a diacylglycerol (DAG) analog. OAG application or increased endogenous DAG, resulting from DAG kinase inhibition, reduced the histamine-induced response. Furthermore, activation of the DAG target, protein kinase C (PKC), by TPA (12-O-tetradecanoyl 4beta-phorbol 13alpha-acetate) resulted in inhibition of the histamine-induced Ca(2+) response, an action prevented by PKC inhibitors. By using reverse transcriptase-polymerase chain reaction analysis, mRNAs for transient receptor potential channels (TRPCs) 1, 4, and 6 as well as for STIM1 (stromal-interacting molecule) and Orai1 were found to be expressed in the U373 MG cells, and confocal microscopy using specific antibodies revealed the presence of the corresponding proteins. Therefore, TRPCs may be candidate proteins forming store-operated channels in the U373 MG cell line. Further, our results confirm the involvement of PKC in the regulation of H(1) receptor-induced responses and point out to the existence of a feedback mechanism acting via PKC to limit the increase in [Ca(2+)](i).
Collapse
Affiliation(s)
- Margarita Barajas
- Departamento de Fisiología, Biofísica y Neurociencias, México, D.F., México
| | | | | | | | | |
Collapse
|
29
|
Narayanan R, Coss CC, Yepuru M, Kearbey JD, Miller DD, Dalton JT. Steroidal androgens and nonsteroidal, tissue-selective androgen receptor modulator, S-22, regulate androgen receptor function through distinct genomic and nongenomic signaling pathways. Mol Endocrinol 2008; 22:2448-65. [PMID: 18801930 DOI: 10.1210/me.2008-0160] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Androgen receptor (AR) ligands are important for the development and function of several tissues and organs. However, the poor oral bioavailability, pharmacokinetic properties, and receptor cross-reactivity of testosterone, coupled with side effects, place limits on its clinical use. Selective AR modulators (SARMs) elicit anabolic effects in muscle and bone, sparing reproductive organs like the prostate. However, molecular mechanisms underlying the tissue selectivity remain ambiguous. We performed a variety of in vitro studies to compare and define the molecular mechanisms of an aryl propionamide SARM, S-22, as compared with dihydrotestosterone (DHT). Studies indicated that S-22 increased levator ani muscle weight but decreased the size of prostate in rats. Analysis of the upstream intracellular signaling events indicated that S-22 and DHT mediated their actions through distinct pathways. Modulation of these pathways altered the recruitment of AR and its cofactors to the PSA enhancer in a ligand-dependent fashion. In addition, S-22 induced Xenopus laevis oocyte maturation and rapid phosphorylation of several kinases, through pathways distinct from steroids. These studies reveal novel differences in the molecular mechanisms by which S-22, a nonsteroidal SARM, and DHT mediate their pharmacological effects.
Collapse
Affiliation(s)
- Ramesh Narayanan
- Preclinical Research and Development, GTx, Inc., 3 North Dunlap Street, Memphis, Tennessee 38163, USA
| | | | | | | | | | | |
Collapse
|
30
|
Muik M, Frischauf I, Derler I, Fahrner M, Bergsmann J, Eder P, Schindl R, Hesch C, Polzinger B, Fritsch R, Kahr H, Madl J, Gruber H, Groschner K, Romanin C. Dynamic coupling of the putative coiled-coil domain of ORAI1 with STIM1 mediates ORAI1 channel activation. J Biol Chem 2008; 283:8014-22. [PMID: 18187424 DOI: 10.1074/jbc.m708898200] [Citation(s) in RCA: 332] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
STIM1 and ORAI1 (also termed CRACM1) are essential components of the classical calcium release-activated calcium current; however, the mechanism of the transmission of information of STIM1 to the calcium release-activated calcium/ORAI1 channel is as yet unknown. Here we demonstrate by Förster resonance energy transfer microscopy a dynamic coupling of STIM1 and ORAI1 that culminates in the activation of Ca(2+) entry. Förster resonance energy transfer imaging of living cells provided insight into the time dependence of crucial events of this signaling pathway comprising Ca(2+) store depletion, STIM1 multimerization, and STIM1-ORAI1 interaction. Accelerated store depletion allowed resolving a significant time lag between STIM1-STIM1 and STIM1-ORAI1 interactions. Store refilling reversed both STIM1 multimerization and STIM1-ORAI1 interaction. The cytosolic STIM1 C terminus itself was able, in vitro as well as in vivo, to associate with ORAI1 and to stimulate channel function, yet without ORAI1-STIM1 cluster formation. The dynamic interaction occurred via the C terminus of ORAI1 that includes a putative coiled-coil domain structure. An ORAI1 C terminus deletion mutant as well as a mutant (L273S) with impeded coiled-coil domain formation lacked both interaction as well as functional communication with STIM1 and failed to generate Ca(2+) inward currents. An N-terminal deletion mutant of ORAI1 as well as the ORAI1 R91W mutant linked to severe combined immune deficiency syndrome was similarly impaired in terms of current activation despite being able to interact with STIM1. Hence, the C-terminal coiled-coil motif of ORAI1 represents a key domain for dynamic coupling to STIM1.
Collapse
Affiliation(s)
- Martin Muik
- Institute of Biophysics, University of Linz, A-4040 Linz, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhao Z, Walczysko P, Zhao M. Intracellular Ca2+ stores are essential for injury induced Ca2+ signaling and re-endothelialization. J Cell Physiol 2007; 214:595-603. [DOI: 10.1002/jcp.21248] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|