51
|
From Stores to Sinks: Structural Mechanisms of Cytosolic Calcium Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 981:215-251. [PMID: 29594864 DOI: 10.1007/978-3-319-55858-5_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
All eukaryotic cells have adapted the use of the calcium ion (Ca2+) as a universal signaling element through the evolution of a toolkit of Ca2+ sensor, buffer and effector proteins. Among these toolkit components, integral and peripheral proteins decorate biomembranes and coordinate the movement of Ca2+ between compartments, sense these concentration changes and elicit physiological signals. These changes in compartmentalized Ca2+ levels are not mutually exclusive as signals propagate between compartments. For example, agonist induced surface receptor stimulation can lead to transient increases in cytosolic Ca2+ sourced from endoplasmic reticulum (ER) stores; the decrease in ER luminal Ca2+ can subsequently signal the opening surface channels which permit the movement of Ca2+ from the extracellular space to the cytosol. Remarkably, the minuscule compartments of mitochondria can function as significant cytosolic Ca2+ sinks by taking up Ca2+ in a coordinated manner. In non-excitable cells, inositol 1,4,5 trisphosphate receptors (IP3Rs) on the ER respond to surface receptor stimulation; stromal interaction molecules (STIMs) sense the ER luminal Ca2+ depletion and activate surface Orai1 channels; surface Orai1 channels selectively permit the movement of Ca2+ from the extracellular space to the cytosol; uptake of Ca2+ into the matrix through the mitochondrial Ca2+ uniporter (MCU) further shapes the cytosolic Ca2+ levels. Recent structural elucidations of these key Ca2+ toolkit components have improved our understanding of how they function to orchestrate precise cytosolic Ca2+ levels for specific physiological responses. This chapter reviews the atomic-resolution structures of IP3R, STIM1, Orai1 and MCU elucidated by X-ray crystallography, electron microscopy and NMR and discusses the mechanisms underlying their biological functions in their respective compartments within the cell.
Collapse
|
52
|
Chandel A, Das KK, Bachhawat AK. Glutathione depletion activates the yeast vacuolar transient receptor potential channel, Yvc1p, by reversible glutathionylation of specific cysteines. Mol Biol Cell 2016; 27:3913-3925. [PMID: 27708136 PMCID: PMC5170613 DOI: 10.1091/mbc.e16-05-0281] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/14/2016] [Accepted: 09/27/2016] [Indexed: 11/12/2022] Open
Abstract
Glutathione depletion leads to calcium influx in yeast cells via plasma membrane Cch1p and the vacuolar Yvc1p channels. Yvc1p, a yeast vacuolar transient receptor potential channel, is activated by glutathionylation carried out by the glutathione S-transferase Gtt1p, and this mechanism is reversible with deglutathionylation being mediated by the thioredoxin Trx2p. Glutathione depletion and calcium influx into the cytoplasm are two hallmarks of apoptosis. We have been investigating how glutathione depletion leads to apoptosis in yeast. We show here that glutathione depletion in yeast leads to the activation of two cytoplasmically inward-facing channels: the plasma membrane, Cch1p, and the vacuolar calcium channel, Yvc1p. Deletion of these channels partially rescues cells from glutathione depletion–induced cell death. Subsequent investigations on the Yvc1p channel, a homologue of the mammalian TRP channels, revealed that the channel is activated by glutathionylation. Yvc1p has nine cysteine residues, of which eight are located in the cytoplasmic regions and one on the transmembrane domain. We show that three of these cysteines, Cys-17, Cys-79, and Cys-191, are specifically glutathionylated. Mutation of these cysteines to alanine leads to a loss in glutathionylation and a concomitant loss in calcium channel activity. We further investigated the mechanism of glutathionylation and demonstrate a role for the yeast glutathione S-transferase Gtt1p in glutathionylation. Yvc1p is also deglutathionylated, and this was found to be mediated by the yeast thioredoxin, Trx2p. A model for redox activation and deactivation of the yeast Yvc1p channel is presented.
Collapse
Affiliation(s)
- Avinash Chandel
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali 140306, Punjab, India
| | - Krishna K Das
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali 140306, Punjab, India
| | - Anand K Bachhawat
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali 140306, Punjab, India
| |
Collapse
|
53
|
Identification of MMP1 as a novel risk factor for intracranial aneurysms in ADPKD using iPSC models. Sci Rep 2016; 6:30013. [PMID: 27418197 PMCID: PMC4945931 DOI: 10.1038/srep30013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/29/2016] [Indexed: 11/08/2022] Open
Abstract
Cardiovascular complications are the leading cause of death in autosomal dominant polycystic kidney disease (ADPKD), and intracranial aneurysm (ICA) causing subarachnoid hemorrhage is among the most serious complications. The diagnostic and therapeutic strategies for ICAs in ADPKD have not been fully established. We here generated induced pluripotent stem cells (iPSCs) from seven ADPKD patients, including four with ICAs. The vascular cells differentiated from ADPKD-iPSCs showed altered Ca(2+) entry and gene expression profiles compared with those of iPSCs from non-ADPKD subjects. We found that the expression level of a metalloenzyme gene, matrix metalloproteinase (MMP) 1, was specifically elevated in iPSC-derived endothelia from ADPKD patients with ICAs. Furthermore, we confirmed the correlation between the serum MMP1 levels and the development of ICAs in 354 ADPKD patients, indicating that high serum MMP1 levels may be a novel risk factor. These results suggest that cellular disease models with ADPKD-specific iPSCs can be used to study the disease mechanisms and to identify novel disease-related molecules or risk factors.
Collapse
|
54
|
Wang JL, Chou CT, Liang WZ, Yeh JH, Kuo CC, Lee CY, Shieh P, Kuo DH, Chen FA, Jan CR. Effect of 2,5-dimethylphenol on Ca(2+) movement and viability in PC3 human prostate cancer cells. Toxicol Mech Methods 2016; 26:327-33. [PMID: 27310574 DOI: 10.3109/15376516.2016.1158893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The phenolic compound 2,5-dimethylphenol is a natural product. 2,5-Dimethylphenol has been shown to affect rat hepatic and pulmonary microsomal metabolism. However, the effect of 2,5-dimethylphenol on Ca(2+ )signaling and cyotoxicity has never been explored in any culture cells. This study explored the effect of 2,5-dimethylphenol on cytosolic free Ca(2+ )levels ([Ca(2+)]i) and cell viability in PC3 human prostate cancer cells. 2,5-Dimethylphenol at concentrations between 500 μM and 1000 μM evoked [Ca(2+)]i rises in a concentration-dependent manner. This Ca(2+ )signal was inhibited by approximately half by the removal of extracellular Ca(2+). 2,5-Dimethylphenol-induced Ca(2+ )influx was confirmed by Mn(2+)-induced quench of fura-2 fluorescence. Pretreatment with the protein kinase C (PKC) inhibitor GF109203X, nifedipine or the store-operated Ca(2+ )entry inhibitors (econazole or SKF96365) inhibited 2,5-dimethylphenol-induced Ca(2+ )signal in Ca(2+)-containing medium by ∼30%. Treatment with the endoplasmic reticulum Ca(2+ )pump inhibitor thapsigargin in Ca(2+)-free medium abolished 2,5-dimethylphenol-induced [Ca(2+)]i rises. Conversely, treatment with 2,5-dimethylphenol abolished thapsigargin-induced [Ca(2+)]i rises. Inhibition of phospholipase C (PLC) with U73122 reduced 2,5-dimethylphenol-evoked [Ca(2+)]i rises by ∼80%. 2,5-Dimethylphenol killed cells at concentrations of 350-1000 μM in a concentration-dependent fashion. Chelation of cytosolic Ca(2+ )with 1,2-bis(2-aminophenoxy)ethane-N, N, N', N'-tetraacetic acid/AM (BAPTA/AM) did not prevent 2,5-dimethylphenol's cytotoxicity. Together, in PC3 cells, 2,5-dimethylphenol induced [Ca(2+)]i rises that involved Ca(2+ )entry through PKC-regulated store-operated Ca(2+ )channels and PLC-dependent Ca(2+ )release from the endoplasmic reticulum. 2,5-Dimethylphenol induced cytotoxicity in a Ca(2+)-independent manner.
Collapse
Affiliation(s)
- Jue-Long Wang
- a Department of Rehabilitation , Kaohsiung Veterans General Hospital Tainan Branch , Tainan , Taiwan , ROC
| | - Chiang-Ting Chou
- b Department of Nursing , Division of Basic Medical Sciences, Chang Gung Institute of Technology , Chia-Yi, Taiwan , ROC .,c Chronic Diseases and Health Promotion Research Center, Chang Gung Institute of Technology , Chia-Yi, Taiwan , ROC
| | - Wei-Zhe Liang
- d Department of Medical Education and Research , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan , ROC
| | - Jeng-Hsien Yeh
- e Department of Pathology and Laboratory Medicine , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan , ROC
| | - Chun-Chi Kuo
- f Department of Nursing , Tzu Hui Institute of Technology , Pingtung , Taiwan , ROC
| | - Chao-Ying Lee
- g School of Pharmacy, China Medical University , Taichung , Taiwan , ROC
| | - Pochuen Shieh
- h Department of Pharmacy , Tajen University , Pingtung , Taiwan , ROC
| | - Daih-Huang Kuo
- h Department of Pharmacy , Tajen University , Pingtung , Taiwan , ROC
| | - Fu-An Chen
- h Department of Pharmacy , Tajen University , Pingtung , Taiwan , ROC
| | - Chung-Ren Jan
- d Department of Medical Education and Research , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan , ROC
| |
Collapse
|
55
|
Hsu SS, Chou CT, Liao WC, Shieh P, Kuo DH, Kuo CC, Jan CR, Liang WZ. The effect of gallic acid on cytotoxicity, Ca(2+) homeostasis and ROS production in DBTRG-05MG human glioblastoma cells and CTX TNA2 rat astrocytes. Chem Biol Interact 2016; 252:61-73. [PMID: 27060209 DOI: 10.1016/j.cbi.2016.04.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/07/2016] [Accepted: 04/05/2016] [Indexed: 01/14/2023]
Abstract
Gallic acid, a polyhydroxylphenolic compound, is widely distributed in various plants, fruits and foods. It has been shown that gallic acid passes into blood brain barrier and reaches the brain tissue of middle cerebral artery occlusion rats. However, the effect of gallic acid on Ca(2+) signaling in glia cells is unknown. This study explored whether gallic acid affected Ca(2+) homeostasis and induced Ca(2+)-associated cytotoxicity in DBTRG-05MG human glioblastoma cells and CTX TNA2 rat astrocytes. Gallic acid (20-40 μM) concentration-dependently induced cytotoxicity and intracellular Ca(2+) level ([Ca(2+)]i) increases in DBTRG-05MG cells but not in CTX TNA2 cells. In DBTRG-05MG cells, the Ca(2+) response was decreased by half by removal of extracellular Ca(2+). In Ca(2+)-containing medium, gallic acid-induced Ca(2+) entry was inhibited by store-operated Ca(2+) channel inhibitors (2-APB, econazole and SKF96365). In Ca(2+)-free medium, pretreatment with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin abolished gallic acid-induced [Ca(2+)]i increases. Conversely, incubation with gallic acid also abolished thapsigargin-induced [Ca(2+)]i increases. Inhibition of phospholipase C with U73122 abolished gallic acid-induced [Ca(2+)]i increases. Gallic acid significantly caused cytotoxicity in DBTRG-05MG cells, which was partially prevented by prechelating cytosolic Ca(2+) with BAPTA-AM. Moreover, gallic acid activated mitochondrial apoptotic pathways that involved ROS production. Together, in DBTRG-05MG cells but not in CTX TNA2 cells, gallic acid induced [Ca(2+)]i increases by causing Ca(2+) entry via 2-APB, econazole and SKF96365-sensitive store-operated Ca(2+) entry, and phospholipase C-dependent release from the endoplasmic reticulum. This Ca(2+) signal subsequently evoked mitochondrial pathways of apoptosis that involved ROS production.
Collapse
Affiliation(s)
- Shu-Shong Hsu
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC; Department of Surgery, National Defense Medical Center, 114 Taipei, Taiwan, ROC
| | - Chiang-Ting Chou
- Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Chia-Yi 613, Taiwan, ROC; Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chia-Yi 613, Taiwan, ROC
| | - Wei-Chuan Liao
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC
| | - Pochuen Shieh
- Department of Pharmacy, Tajen University, Pingtung 907, Taiwan, ROC
| | - Daih-Huang Kuo
- Department of Pharmacy, Tajen University, Pingtung 907, Taiwan, ROC
| | - Chun-Chi Kuo
- Department of Nursing, Tzu Hui Institute of Technology, Pingtung 907, Taiwan, ROC
| | - Chung-Ren Jan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan, ROC
| | - Wei-Zhe Liang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan, ROC.
| |
Collapse
|
56
|
Endostatin is protective against monocrotaline-induced right heart disease through the inhibition of T-type Ca(2+) channel. Pflugers Arch 2016; 468:1259-1270. [PMID: 27023352 DOI: 10.1007/s00424-016-1810-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/03/2016] [Accepted: 03/15/2016] [Indexed: 12/27/2022]
Abstract
Endostatin (ES), a C-terminal fragment of collagen XVIIIα1, has a potent anti-angiogenic effect. ES prevents tumor proliferation through inhibiting T-type Ca(2+) channel. T-type Ca(2+) channel is re-expressed during heart diseases including monocrotaline (MCT)-induced right heart failure. The present study aimed to clarify the effects of ES on T-type Ca(2+) channel and pathogenesis of MCT-induced right ventricular disease. MCT or saline was injected intraperitoneally to rats. After cardiomyocytes were isolated from right ventricles (RVs), T-type Ca(2+) channel current (I CaT) was measured by a patch-clamp method. After ES small interfering RNA (siRNA) or control siRNA (20 μg) was administrated for 1 week via the right jugular vein 1 week after MCT injection, echocardiography and histological analysis were done. I CaT was significantly increased in RV from MCT-injected rats, and ES significantly inhibited it. The survival rate of ES siRNA-administrated MCT rats (MCT ES si group) was decreased. In echocardiography, although ES siRNA did not affect pulmonary arterial pressure, RV systolic function was impaired in MCT ES si group compared with control siRNA-administrated MCT rats (MCT cont si group). In the histological analysis of RV, ES expression was increased in MCT cont si group, and ES siRNA inhibited it. Furthermore, although MCT cont si group showed only cardiomyocyte hypertrophy, MCT ES si group showed notable enlargement of intercellular spaces. The present study for the first time revealed that ES inhibits T-type Ca(2+) channel activity in RV from MCT-injected rats. ES gene knockdown deteriorates MCT-induced right heart disease. ES is thus cardioprotective possibly through inhibiting T-type Ca(2+) channel activity.
Collapse
|
57
|
Bcl-2 proteins and calcium signaling: complexity beneath the surface. Oncogene 2016; 35:5079-92. [DOI: 10.1038/onc.2016.31] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 12/12/2022]
|
58
|
Cao Z, Liu D, Zhang Q, Sun X, Li Y. Aluminum Chloride Induces Osteoblasts Apoptosis via Disrupting Calcium Homeostasis and Activating Ca(2+)/CaMKII Signal Pathway. Biol Trace Elem Res 2016; 169:247-53. [PMID: 26138010 DOI: 10.1007/s12011-015-0417-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/17/2015] [Indexed: 12/16/2022]
Abstract
Aluminum promotes osteoblast (OB) apoptosis. Apoptosis is induced by the disordered calcium homeostasis. Therefore, to investigate the relationship between Al-induced OB apoptosis and calcium homeostasis, calvarium OBs from neonatal rats (3-4 days) were cultured and exposed to 0.048-mg/mL Al(3+) or 0.048-mg/mL Al(3+) combined with 5 μM BAPTA-AM (OBs were pretreated with 5 μM BAPTA-AM for 1 h, then added 0.048 mg/mL Al(3+)), respectively. Then OB apoptosis rate, intracellular calcium ions concentration ([Ca(2+)]i), mRNA expression level of calmodulin (CaM), and protein expression levels of CaM and p-CaMKII in OBs were examined. The result showed that AlCl3 increased OB apoptosis rate, and [Ca(2+)]i and p-CaMKII expression levels and decreased CaM expression levels, whereas BAPTA-AM relieved the effects. These results proved that AlCl3 induced OB apoptosis by disrupting the intracellular Ca(2+) homeostasis and activating the Ca(2+)/CaMKII signal pathway. Our findings can provide new insights for revealing the apoptosis mechanism of OBs exposed to AlCl3.
Collapse
Affiliation(s)
- Zheng Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Dawei Liu
- Heilongjiang Province Hospital, Harbin, 150036, China
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Qiuyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xudong Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yanfei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
59
|
Croft W, Reusch K, Tilunaite A, Russell NA, Thul R, Bellamy TC. Probabilistic encoding of stimulus strength in astrocyte global calcium signals. Glia 2015; 64:537-52. [PMID: 26651126 DOI: 10.1002/glia.22947] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022]
Abstract
Astrocyte calcium signals can range in size from subcellular microdomains to waves that spread through the whole cell (and into connected cells). The differential roles of such local or global calcium signaling are under intense investigation, but the mechanisms by which local signals evolve into global signals in astrocytes are not well understood, nor are the computational rules by which physiological stimuli are transduced into a global signal. To investigate these questions, we transiently applied receptor agonists linked to calcium signaling to primary cultures of cerebellar astrocytes. Astrocytes repetitively tested with the same stimulus responded with global signals intermittently, indicating that each stimulus had a defined probability for triggering a response. The response probability varied between agonists, increased with agonist concentration, and could be positively and negatively modulated by crosstalk with other signaling pathways. To better understand the processes determining the evolution of a global signal, we recorded subcellular calcium "puffs" throughout the whole cell during stimulation. The key requirement for puffs to trigger a global calcium wave following receptor activation appeared to be the synchronous release of calcium from three or more sites, rather than an increasing calcium load accumulating in the cytosol due to increased puff size, amplitude, or frequency. These results suggest that the concentration of transient stimuli will be encoded into a probability of generating a global calcium response, determined by the likelihood of synchronous release from multiple subcellular sites.
Collapse
Affiliation(s)
- Wayne Croft
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Katharina Reusch
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.,Department of Electrical and Electronic Engineering, University of Nottingham, Nottingham, United Kingdom
| | - Agne Tilunaite
- School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Noah A Russell
- Department of Electrical and Electronic Engineering, University of Nottingham, Nottingham, United Kingdom
| | - Rüdiger Thul
- School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Tomas C Bellamy
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
60
|
Cui X, Luo Y, Li C, Li Y, Wang Z. Changes of intracellular Ca2+ in quercetin-induced autophagy progression. Acta Biochim Biophys Sin (Shanghai) 2015; 47:908-14. [PMID: 26423114 DOI: 10.1093/abbs/gmv096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/29/2015] [Indexed: 12/22/2022] Open
Abstract
Quercetin was previously reported to exhibit significant anti-proliferative activities, and its major effect on tumors was to induce cell apoptosis or autophagy. However, the specific mechanism remains controversial. In this study, autophagy induced by quercetin was determined with various methods. Intracellular Ca2+ ([Ca2+]i) was measured after being incubated with Fluo-3 acetoxymethyl (AM). At the same time, the relationship between the intracellular Ca2+ and autophagy induced by quercetin was further analyzed. These results showed that autophagy induced by quercetin (0-50 µg/ml) in HepG2 cells was in a dose-dependent manner. Meanwhile, when autophagy was induced by quercetin, [Ca2+]i was significantly increased. And after being incubated with calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N-tetraacetic acid-AM, autophagy was suppressed, which implied that [Ca2+]i elevation appeared to be the cause for autophagy induction. These results suggested that calcium from intracellular calcium storage may play an important role in quercetin-induced autophagy.
Collapse
Affiliation(s)
- Xiaodong Cui
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Yanan Luo
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Chen Li
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Yuying Li
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Zhuanhua Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
61
|
Chung HK, Rathor N, Wang SR, Wang JY, Rao JN. RhoA enhances store-operated Ca2+ entry and intestinal epithelial restitution by interacting with TRPC1 after wounding. Am J Physiol Gastrointest Liver Physiol 2015; 309:G759-67. [PMID: 26336927 PMCID: PMC4628965 DOI: 10.1152/ajpgi.00185.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/26/2015] [Indexed: 01/31/2023]
Abstract
Early mucosal restitution occurs as a consequence of epithelial cell migration to resealing of superficial wounds after injury. Our previous studies show that canonical transient receptor potential-1 (TRPC1) functions as a store-operated Ca(2+) channel (SOC) in intestinal epithelial cells (IECs) and plays an important role in early epithelial restitution by increasing Ca(2+) influx. Here we further reported that RhoA, a small GTP-binding protein, interacts with and regulates TRPC1, thus enhancing SOC-mediated Ca(2+) entry (SOCE) and epithelial restitution after wounding. RhoA physically associated with TRPC1 and formed the RhoA/TRPC1 complexes, and this interaction increased in stable TRPC1-transfected IEC-6 cells (IEC-TRPC1). Inactivation of RhoA by treating IEC-TRPC1 cells with exoenzyme C3 transferase (C3) or ectopic expression of dominant negative RhoA (DNMRhoA) reduced RhoA/TRPC1 complexes and inhibited Ca(2+) influx after store depletion, which was paralleled by an inhibition of cell migration over the wounded area. In contrast, ectopic expression of wild-type (WT)-RhoA increased the levels of RhoA/TRPC1 complexes, induced Ca(2+) influx through activation of SOCE, and promoted cell migration after wounding. TRPC1 silencing by transfecting stable WT RhoA-transfected cells with siRNA targeting TRPC1 (siTRPC1) reduced SOCE and repressed epithelial restitution. Moreover, ectopic overexpression of WT-RhoA in polyamine-deficient cells rescued the inhibition of Ca(2+) influx and cell migration induced by polyamine depletion. These findings indicate that RhoA interacts with and activates TRPC1 and thus stimulates rapid epithelial restitution after injury by inducing Ca(2+) signaling.
Collapse
Affiliation(s)
- Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Navneeta Rathor
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Shelley R Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland; and Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
62
|
Wolf IMA, Diercks BP, Gattkowski E, Czarniak F, Kempski J, Werner R, Schetelig D, Mittrücker HW, Schumacher V, von Osten M, Lodygin D, Flügel A, Fliegert R, Guse AH. Frontrunners of T cell activation: Initial, localized Ca2+ signals mediated by NAADP and the type 1 ryanodine receptor. Sci Signal 2015; 8:ra102. [PMID: 26462735 DOI: 10.1126/scisignal.aab0863] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The activation of T cells is the fundamental on switch for the adaptive immune system. Ca(2+) signaling is essential for T cell activation and starts as initial, short-lived, localized Ca(2+) signals. The second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) forms rapidly upon T cell activation and stimulates early Ca(2+) signaling. We developed a high-resolution imaging technique using multiple fluorescent Ca(2+) indicator dyes to characterize these early signaling events and investigate the channels involved in NAADP-dependent Ca(2+) signals. In the first seconds of activation of either primary murine T cells or human Jurkat cells with beads coated with an antibody against CD3, we detected Ca(2+) signals with diameters close to the limit of detection and that were close to the activation site at the plasma membrane. In Jurkat cells in which the ryanodine receptor (RyR) was knocked down or in primary T cells from RyR1(-/-) mice, either these early Ca(2+) signals were not detected or the number of signals was markedly reduced. Local Ca(2+) signals observed within 20 ms upon microinjection of Jurkat cells with NAADP were also sensitive to RyR knockdown. In contrast, TRPM2 (transient receptor potential channel, subtype melastatin 2), a potential NAADP target channel, was not required for the formation of initial Ca(2+) signals in primary T cells. Thus, through our high-resolution imaging method, we characterized early Ca(2+) release events in T cells and obtained evidence for the involvement of RyR and NAADP in such signals.
Collapse
Affiliation(s)
- Insa M A Wolf
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ellen Gattkowski
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Frederik Czarniak
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jan Kempski
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - René Werner
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Daniel Schetelig
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Valéa Schumacher
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Manuel von Osten
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Dimitri Lodygin
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Alexander Flügel
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Ralf Fliegert
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
63
|
Stavermann M, Meuth P, Doengi M, Thyssen A, Deitmer JW, Lohr C. Calcium-induced calcium release and gap junctions mediate large-scale calcium waves in olfactory ensheathing cells in situ. Cell Calcium 2015; 58:215-25. [DOI: 10.1016/j.ceca.2015.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/04/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023]
|
64
|
Ross CL, Siriwardane M, Almeida-Porada G, Porada CD, Brink P, Christ GJ, Harrison BS. The effect of low-frequency electromagnetic field on human bone marrow stem/progenitor cell differentiation. Stem Cell Res 2015; 15:96-108. [PMID: 26042793 PMCID: PMC4516580 DOI: 10.1016/j.scr.2015.04.009] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/17/2015] [Accepted: 04/27/2015] [Indexed: 12/12/2022] Open
Abstract
Human bone marrow stromal cells (hBMSCs, also known as bone marrow-derived mesenchymal stem cells) are a population of progenitor cells that contain a subset of skeletal stem cells (hSSCs), able to recreate cartilage, bone, stroma that supports hematopoiesis and marrow adipocytes. As such, they have become an important resource in developing strategies for regenerative medicine and tissue engineering due to their self-renewal and differentiation capabilities. The differentiation of SSCs/BMSCs is dependent on exposure to biophysical and biochemical stimuli that favor early and rapid activation of the in vivo tissue repair process. Exposure to exogenous stimuli such as an electromagnetic field (EMF) can promote differentiation of SSCs/BMSCs via ion dynamics and small signaling molecules. The plasma membrane is often considered to be the main target for EMF signals and most results point to an effect on the rate of ion or ligand binding due to a receptor site acting as a modulator of signaling cascades. Ion fluxes are closely involved in differentiation control as stem cells move and grow in specific directions to form tissues and organs. EMF affects numerous biological functions such as gene expression, cell fate, and cell differentiation, but will only induce these effects within a certain range of low frequencies as well as low amplitudes. EMF has been reported to be effective in the enhancement of osteogenesis and chondrogenesis of hSSCs/BMSCs with no documented negative effects. Studies show specific EMF frequencies enhance hSSC/BMSC adherence, proliferation, differentiation, and viability, all of which play a key role in the use of hSSCs/BMSCs for tissue engineering. While many EMF studies report significant enhancement of the differentiation process, results differ depending on the experimental and environmental conditions. Here we review how specific EMF parameters (frequency, intensity, and time of exposure) significantly regulate hSSC/BMSC differentiation in vitro. We discuss optimal conditions and parameters for effective hSSC/BMSC differentiation using EMF treatment in an in vivo setting, and how these can be translated to clinical trials.
Collapse
Affiliation(s)
- Christina L Ross
- Wake Forest Institute for Regenerative Medicine, USA; Wake Forest Center for Integrative Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | | | | | | | - Peter Brink
- Department of Physiology and Biophysics, SUNY Stony Brook, Stony Brook, NY 11794, USA
| | | | | |
Collapse
|
65
|
Chang HT, Chou CT, Yu CC, Tsai JY, Sun TK, Liang WZ, Lin KL, Tseng HW, Kuo CC, Chen FA, Kuo DH, Pan CC, Ho CM, Shieh P, Jan CR. The mechanism of protriptyline-induced Ca2+movement and non-Ca2+-triggered cell death in PC3 human prostate cancer cells. J Recept Signal Transduct Res 2015; 35:429-34. [DOI: 10.3109/10799893.2014.1000464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
66
|
Suzuki Y, Yokoyama K. Development of Functional Fluorescent Molecular Probes for the Detection of Biological Substances. BIOSENSORS 2015; 5:337-63. [PMID: 26095660 PMCID: PMC4493553 DOI: 10.3390/bios5020337] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 01/27/2023]
Abstract
This review is confined to sensors that use fluorescence to transmit biochemical information. Fluorescence is, by far, the most frequently exploited phenomenon for chemical sensors and biosensors. Parameters that define the application of such sensors include intensity, decay time, anisotropy, quenching efficiency, and luminescence energy transfer. To achieve selective (bio)molecular recognition based on these fluorescence phenomena, various fluorescent elements such as small organic molecules, enzymes, antibodies, and oligonucleotides have been designed and synthesized over the past decades. This review describes the immense variety of fluorescent probes that have been designed for the recognitions of ions, small and large molecules, and their biological applications in terms of intracellular fluorescent imaging techniques.
Collapse
Affiliation(s)
- Yoshio Suzuki
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba 305-8566, Japan.
| | - Kenji Yokoyama
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1 Katakura, Hachioji, Tokyo 192-0982, Japan.
| |
Collapse
|
67
|
Frequency and relative prevalence of calcium blips and puffs in a model of small IP₃R clusters. Biophys J 2015; 106:2353-63. [PMID: 24896114 DOI: 10.1016/j.bpj.2014.04.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 03/07/2014] [Accepted: 04/15/2014] [Indexed: 11/22/2022] Open
Abstract
In this work, we model the local calcium release from clusters with a few inositol 1,4,5-trisphosphate receptor (IP3R) channels, focusing on the stochastic process in which an open channel either triggers other channels to open (as a puff) or fails to cause any channel to open (as a blip). We show that there are linear relations for the interevent interval (including blips and puffs) and the first event latency against the inverse cluster size. However, nonlinearity is found for the interpuff interval and the first puff latency against the inverse cluster size. Furthermore, the simulations indicate that the blip fraction among all release events and the blip frequency are increasing with larger basal [Ca(2+)], with blips in turn giving a growing contribution to basal [Ca(2+)]. This result suggests that blips are not just lapses to trigger puffs, but they may also possess a biological function to contribute to the initiation of calcium waves by a preceding increase of basal [Ca(2+)] in cells that have small IP3R clusters.
Collapse
|
68
|
Rückl M, Parker I, Marchant JS, Nagaiah C, Johenning FW, Rüdiger S. Modulation of elementary calcium release mediates a transition from puffs to waves in an IP3R cluster model. PLoS Comput Biol 2015; 11:e1003965. [PMID: 25569772 PMCID: PMC4288706 DOI: 10.1371/journal.pcbi.1003965] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/06/2014] [Indexed: 11/18/2022] Open
Abstract
The oscillating concentration of intracellular calcium is one of the most important examples for collective dynamics in cell biology. Localized releases of calcium through clusters of inositol 1,4,5-trisphosphate receptor channels constitute elementary signals called calcium puffs. Coupling by diffusing calcium leads to global releases and waves, but the exact mechanism of inter-cluster coupling and triggering of waves is unknown. To elucidate the relation of puffs and waves, we here model a cluster of IP3R channels using a gating scheme with variable non-equilibrium IP3 binding. Hybrid stochastic and deterministic simulations show that puffs are not stereotyped events of constant duration but are sensitive to stimulation strength and residual calcium. For increasing IP3 concentration, the release events become modulated at a timescale of minutes, with repetitive wave-like releases interspersed with several puffs. This modulation is consistent with experimental observations we present, including refractoriness and increase of puff frequency during the inter-wave interval. Our results suggest that waves are established by a random but time-modulated appearance of sustained release events, which have a high potential to trigger and synchronize activity throughout the cell.
Collapse
Affiliation(s)
- Martin Rückl
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ian Parker
- Departments of Neurobiology and Behavior, Physiology and Biophysics, University of California, Irvine, Irvine, California, United States of America
| | - Jonathan S. Marchant
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Chamakuri Nagaiah
- Johann Radon Institute for Computational and Applied Mathematics, Austrian Academy of Sciences, Linz, Austria
| | | | - Sten Rüdiger
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
69
|
Graber ZT, Wang W, Singh G, Kuzmenko I, Vaknin D, Kooijman EE. Competitive cation binding to phosphatidylinositol-4,5-bisphosphate domains revealed by X-ray fluorescence. RSC Adv 2015. [DOI: 10.1039/c5ra19023a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Calcium ions bind strongly to PIP2 at physiological concentrations, leading to condensation and decreased effective charge for PIP2. Calcium displaces the more numerous magnesium and potassium ions, but some potassium ions remain.
Collapse
Affiliation(s)
- Z. T. Graber
- Department of Chemistry and Biochemistry
- Kent State University
- Kent
- USA
| | - W. Wang
- Ames Laboratory and Department of Physics and Astronomy
- Iowa State University
- Ames
- USA
| | - G. Singh
- Department of Physics
- Kent State University
- Kent
- USA
| | - I. Kuzmenko
- X-ray Science Division
- Advanced Photon Source
- Argonne National Laboratory
- Lemont
- USA
| | - D. Vaknin
- Ames Laboratory and Department of Physics and Astronomy
- Iowa State University
- Ames
- USA
| | - E. E. Kooijman
- Department of Biological Sciences
- Kent State University
- Kent
- USA
| |
Collapse
|
70
|
Liang WZ, Chou CT, Chang HT, Cheng JS, Kuo DH, Ko KC, Chiang NN, Wu RF, Shieh P, Jan CR. The mechanism of honokiol-induced intracellular Ca(2+) rises and apoptosis in human glioblastoma cells. Chem Biol Interact 2014; 221:13-23. [PMID: 25106108 DOI: 10.1016/j.cbi.2014.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 07/15/2014] [Accepted: 07/25/2014] [Indexed: 12/21/2022]
Abstract
Honokiol, an active constituent of oriental medicinal herb Magnolia officinalis, caused Ca(2+) mobilization and apoptosis in different cancer cells. In vivo, honokiol crossed the blood-brain or -cerebrospinal fluid barrier, suggesting that it may be an effective drug for the treatment of brain tumors, including glioblastoma. This study examined the effect of honokiol on intracellular Ca(2+) concentration ([Ca(2+)]i) and apoptosis in DBTRG-05MG human glioblastoma cells. Honokiol concentration-dependently induced a [Ca(2+)]i rise. The signal was decreased partially by removal of extracellular Ca(2+). Honokiol-triggered [Ca(2+)]i rise was not suppressed by store-operated Ca(2+) channel blockers (nifedipine, econazole, SK&F96365) and the protein kinase C (PKC) activator phorbol 12-myristate 13 acetate (PMA), but was inhibited by the PKC inhibitor GF109203X. GF109203X-induced inhibition was not altered by removal of extracellular Ca(2+). In Ca(2+)-free medium, pretreatment with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin (TG) or 2,5-di-tert-butylhydroquinone (BHQ) abolished honokiol-induced [Ca(2+)]i rise. Conversely, incubation with honokiol abolished TG or BHQ-induced [Ca(2+)]i rise. Inhibition of phospholipase C (PLC) with U73122 abolished honokiol-induced [Ca(2+)]i rise. Honokiol (20-80μM) reduced the cell viability, which was not reversed by prechelating cytosolic Ca(2+) with BAPTA-AM (1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester). Honokiol (20-60μM) enhanced reactive oxygen species (ROS) production, decreased mitochondrial membrane potential, released cytochrome c, and activated caspase-9/caspase-3. Together, honokiol induced a [Ca(2+)]i rise by inducing PLC-dependent Ca(2+) release from the endoplasmic reticulum and Ca(2+) entry via PKC-dependent, non store-operated Ca(2+) channels. Moreover, honokiol activated the mitochondrial pathway of apoptosis in DBTRG-05MG human glioblastoma cells.
Collapse
Affiliation(s)
- Wei-Zhe Liang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC
| | - Chiang-Ting Chou
- Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Chia-Yi 613, Taiwan, ROC; Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chia-Yi 613, Taiwan, ROC
| | - Hong-Tai Chang
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC
| | - Jin-Shiung Cheng
- Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC
| | - Daih-Huang Kuo
- Department of Pharmacy, Tajen University, Pingtung 907, Taiwan, ROC
| | - Kuang-Chung Ko
- Department of Gastroenterology, Kaohsiung Veterans General Hospital-Pingtung Branch 912, Taiwan, ROC
| | - Ni-Na Chiang
- Department of Pharmacy, Kaohsiung Veterans General Hospital-Pingtung Branch 912, Taiwan, ROC
| | - Ru-Fang Wu
- Department of Pharmacy, Kaohsiung Municipal Kai-Syuan Psychiatric Hospital, Kaohsiung 802, Taiwan, ROC
| | - Pochuen Shieh
- Department of Pharmacy, Tajen University, Pingtung 907, Taiwan, ROC
| | - Chung-Ren Jan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC.
| |
Collapse
|
71
|
Amcheslavsky A, Safrina O, Cahalan MD. Orai3 TM3 point mutation G158C alters kinetics of 2-APB-induced gating by disulfide bridge formation with TM2 C101. ACTA ACUST UNITED AC 2014; 142:405-12. [PMID: 24081982 PMCID: PMC3787773 DOI: 10.1085/jgp.201311030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
After endoplasmic reticulum (ER) Ca2+ store depletion, Orai channels in the plasma membrane (PM) are activated directly by ER-resident STIM proteins to form the Ca2+-selective Ca2+ release–activated Ca2+ (CRAC) channel. However, in the absence of Ca2+ store depletion and STIM interaction, the mammalian homologue Orai3 can be activated by 2-aminoethyl diphenylborinate (2-APB), resulting in a nonselective cation conductance characterized by biphasic inward and outward rectification. Here, we use site-directed mutagenesis and patch-clamp analysis to better understand the mechanism by which 2-APB activates Orai3. We find that point mutation of glycine 158 in the third transmembrane (TM) segment to cysteine, but not alanine, slows the kinetics of 2-APB activation and prevents complete channel closure upon 2-APB washout. The “slow” phenotype exhibited by Orai3 mutant G158C reveals distinct open states, characterized by variable reversal potentials. The slow phenotype can be reversed by application of the reducing reagent bis(2-mercaptoethylsulfone) (BMS), but in a state-dependent manner, only during 2-APB activation. Moreover, the double mutant C101G/G158C, in which an endogenous TM2 cysteine is changed to glycine, does not exhibit altered kinetics of 2-APB activation. We suggest that a disulfide bridge, formed between the introduced cysteine at TM3 position 158 and the endogenous cysteine at TM2 position 101, hinders transitions between Orai3 open and closed states. Our data provide functional confirmation of the proximity of these two residues and suggest a location within the Orai3 protein that is sensitive to the actions of 2-APB.
Collapse
Affiliation(s)
- Anna Amcheslavsky
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697
| | | | | |
Collapse
|
72
|
Rathor N, Zhuang R, Wang JY, Donahue JM, Turner DJ, Rao JN. Src-mediated caveolin-1 phosphorylation regulates intestinal epithelial restitution by altering Ca(2+) influx after wounding. Am J Physiol Gastrointest Liver Physiol 2014; 306:G650-8. [PMID: 24557763 PMCID: PMC3989706 DOI: 10.1152/ajpgi.00003.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Early mucosal restitution occurs as a consequence of intestinal epithelial cell (IEC) migration to reseal superficial wounds, but its exact mechanism remains largely unknown. Caveolin-1 (Cav1), a major component associated with caveolar lipid rafts in the plasma membrane, is implicated in many aspects of cellular functions. This study determined if c-Src kinase (Src)-induced Cav1 phosphorylation promotes intestinal epithelial restitution after wounding by activating Cav1-mediated Ca(2+) signaling. Src directly interacted with Cav1, formed Cav1-Src complexes, and phosphorylated Cav1 in IECs. Inhibition of Src activity by its chemical inhibitor PP2 or suppression of the functional caveolin scaffolding domain by caveolin-scaffolding domain peptides prevented Cav1-Src interaction, reduced Cav1 phosphorylation, decreased Ca(2+) influx, and inhibited cell migration after wounding. Disruption of caveolar lipid raft microdomains by methyl-β-cyclodextrin reduced Cav1-mediated Ca(2+) influx and repressed epithelial restitution. Moreover, Src silencing prevented subcellular redistribution of phosphorylated Cav1 in migrating IECs. These results indicate that Src-induced Cav1 phosphorylation stimulates epithelial restitution by increasing Cav1-mediated Ca(2+) signaling after wounding, thus contributing to the maintenance of gut mucosal integrity under various pathological conditions.
Collapse
Affiliation(s)
- Navneeta Rathor
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Ran Zhuang
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Jian-Ying Wang
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and ,3Department of Pathology, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland
| | - James M. Donahue
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Douglas J. Turner
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| | - Jaladanki N. Rao
- 1Department of Surgery, Cell Biology Group, University of Maryland School of Medicine, Baltimore, Maryland; ,2Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; and
| |
Collapse
|
73
|
Amcheslavsky A, Safrina O, Cahalan MD. State-dependent block of Orai3 TM1 and TM3 cysteine mutants: insights into 2-APB activation. ACTA ACUST UNITED AC 2014; 143:621-31. [PMID: 24733836 PMCID: PMC4003185 DOI: 10.1085/jgp.201411171] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Residue E165, in transmembrane helix 3, participates in formation of the dilated pore of the 2-APB–activated Orai3 channel but not that of the more selective store-operated Orai3 pore. After endoplasmic reticulum (ER) Ca2+ store depletion, Orai channels in the plasma membrane (PM) are activated directly by ER-resident stromal interacting molecule (STIM) proteins to form the Ca2+-selective Ca2+ release-activated Ca2+ (CRAC) channel. Of the three human Orai channel homologues, only Orai3 can be activated by high concentrations (>50 µM) of 2-aminoethyl diphenylborinate (2-APB). 2-APB activation of Orai3 occurs without STIM1–Orai3 interaction or store depletion, and results in a cationic, nonselective current characterized by biphasic inward and outward rectification. Here we use cysteine scanning mutagenesis, thiol-reactive reagents, and patch-clamp analysis to define the residues that assist in formation of the 2-APB–activated Orai3 pore. Mutating transmembrane (TM) 1 residues Q83, V77, and L70 to cysteine results in potentiated block by cadmium ions (Cd2+). TM1 mutants E81C, G73A, G73C, and R66C form channels that are not sensitive to 2-APB activation. We also find that Orai3 mutant V77C is sensitive to block by 2-aminoethyl methanethiosulfonate (MTSEA), but not 2-(trimethylammonium)ethyl methanethiosulfonate (MTSET). Block induced by reaction with MTSEA is state dependent, as it occurs only when Orai3-V77C channels are opened by either 2-APB or by cotransfection with STIM1 and concurrent passive store depletion. We also analyzed TM3 residue E165. Mutation E165A in Orai3 results in diminished 2-APB–activated currents. However, it has little effect on store-operated current density. Furthermore, mutation E165C results in Cd2+-induced block that is state dependent: Cd2+ only blocks 2-APB–activated, not store-operated, mutant channels. Our data suggest that the dilated pore of 2-APB–activated Orai3 is lined by TM1 residues, but also allows for TM3 E165 to approach the central axis of the channel that forms the conducting pathway, or pore.
Collapse
Affiliation(s)
- Anna Amcheslavsky
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697
| | | | | |
Collapse
|
74
|
Hashimoto M, Nara T, Hirawake H, Morales J, Enomoto M, Mikoshiba K. Antisense oligonucleotides targeting parasite inositol 1,4,5-trisphosphate receptor inhibits mammalian host cell invasion by Trypanosoma cruzi. Sci Rep 2014; 4:4231. [PMID: 24577136 PMCID: PMC3937783 DOI: 10.1038/srep04231] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 02/06/2014] [Indexed: 11/17/2022] Open
Abstract
Chagas disease is caused by an intracellular parasitic protist, Trypanosoma cruzi. As there are no highly effective drugs against this agent that also demonstrate low toxicity, there is an urgent need for development of new drugs to treat Chagas disease. We have previously demonstrated that the parasite inositol 1,4,5-trisphosphate receptor (TcIP3R) is crucial for invasion of the mammalian host cell by T. cruzi. Here, we report that TcIP3R is a short-lived protein and that its expression is significantly suppressed in trypomastigotes. Treatment of trypomastigotes, an infective stage of T. cruzi, with antisense oligonucleotides specific to TcIP3R deceased TcIP3R protein levels and impaired trypomastigote invasion of host cells. Due to the resulting instability and very low expression level of TcIP3R in trypomastigotes indicates that TcIP3R is a promising target for antisense therapy in Chagas disease.
Collapse
Affiliation(s)
- Muneaki Hashimoto
- Department of Molecular and Cellular Parasitology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Takeshi Nara
- Department of Molecular and Cellular Parasitology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hiroko Hirawake
- Department of Molecular and Cellular Parasitology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Jorge Morales
- Department of Molecular and Cellular Parasitology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masahiro Enomoto
- Division of Signaling Biology, Ontario Cancer Institute, Toronto, ON, Canada M5G 1L7
| | - Katsuhiko Mikoshiba
- 1] Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Saitama 351-0198, Japan [2] Calcium Oscillation Project, International Cooperative Research Project and Solution-Oriented Research for Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
75
|
Prevarskaya N, Ouadid-Ahidouch H, Skryma R, Shuba Y. Remodelling of Ca2+ transport in cancer: how it contributes to cancer hallmarks? Philos Trans R Soc Lond B Biol Sci 2014; 369:20130097. [PMID: 24493745 DOI: 10.1098/rstb.2013.0097] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer involves defects in the mechanisms underlying cell proliferation, death and migration. Calcium ions are central to these phenomena, serving as major signalling agents with spatial localization, magnitude and temporal characteristics of calcium signals ultimately determining cell's fate. Cellular Ca(2+) signalling is determined by the concerted action of a molecular Ca(2+)-handling toolkit which includes: active energy-dependent Ca(2+) transporters, Ca(2+)-permeable ion channels, Ca(2+)-binding and storage proteins, Ca(2+)-dependent effectors. In cancer, because of mutations, aberrant expression, regulation and/or subcellular targeting of Ca(2+)-handling/transport protein(s) normal relationships among extracellular, cytosolic, endoplasmic reticulum and mitochondrial Ca(2+) concentrations or spatio-temporal patterns of Ca(2+) signalling become distorted. This causes deregulation of Ca(2+)-dependent effectors that control signalling pathways determining cell's behaviour in a way to promote pathophysiological cancer hallmarks such as enhanced proliferation, survival and invasion. Despite the progress in our understanding of Ca(2+) homeostasis remodelling in cancer cells as well as in identification of the key Ca(2+)-transport molecules promoting certain malignant phenotypes, there is still a lot of work to be done to transform fundamental findings and concepts into new Ca(2+) transport-targeting tools for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Natalia Prevarskaya
- Inserm, U1003, Laboratoire de Physiologie Cellulaire, Equipe labellisée par la Ligue contre le cancer, Villeneuve d'Ascq, F-59650 France; Laboratory of Excellence, Ion Channels Science and Therapeutics; Universite de Lille 1, , Villeneuve d'Ascq, F-59650 France
| | | | | | | |
Collapse
|
76
|
Fedorenko OA, Popugaeva E, Enomoto M, Stathopulos PB, Ikura M, Bezprozvanny I. Intracellular calcium channels: inositol-1,4,5-trisphosphate receptors. Eur J Pharmacol 2013; 739:39-48. [PMID: 24300389 DOI: 10.1016/j.ejphar.2013.10.074] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 09/28/2013] [Accepted: 10/17/2013] [Indexed: 01/13/2023]
Abstract
The inositol-1,4,5-trisphosphate receptors (InsP3Rs) are the major intracellular Ca(2+)-release channels in cells. Activity of InsP3Rs is essential for elementary and global Ca(2+) events in the cell. There are three InsP3Rs isoforms that are present in mammalian cells. In this review we will focus primarily on InsP3R type 1. The InsP3R1 is a predominant isoform in neurons and it is the most extensively studied isoform. Combination of biophysical and structural methods revealed key mechanisms of InsP3R function and modulation. Cell biological and biochemical studies lead to identification of a large number of InsP3R-binding proteins. InsP3Rs are involved in the regulation of numerous physiological processes, including learning and memory, proliferation, differentiation, development and cell death. Malfunction of InsP3R1 play a role in a number of neurodegenerative disorders and other disease states. InsP3Rs represent a potentially valuable drug target for treatment of these disorders and for modulating activity of neurons and other cells. Future studies will provide better understanding of physiological functions of InsP3Rs in health and disease.
Collapse
Affiliation(s)
- Olena A Fedorenko
- Department of Brain Physiology, Bogomoletz Institute of Physiology, 01024 Kiev, Ukraine; State Key Laboratory of Molecular and Cellular Biology, 01024 Kiev, Ukraine
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Department of Medical Physics, St. Petersburg State Polytechnical University, 195251 St. Petersburg, Russia
| | - Masahiro Enomoto
- Princess Margaret Cancer Centre, Department of Medical Biophysics, University of Toronto, M5G1L7 Toronto, Ontario, Canada
| | - Peter B Stathopulos
- Princess Margaret Cancer Centre, Department of Medical Biophysics, University of Toronto, M5G1L7 Toronto, Ontario, Canada
| | - Mitsuhiko Ikura
- Princess Margaret Cancer Centre, Department of Medical Biophysics, University of Toronto, M5G1L7 Toronto, Ontario, Canada
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Department of Medical Physics, St. Petersburg State Polytechnical University, 195251 St. Petersburg, Russia; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
77
|
Abstract
Local Ca(2+) signals through voltage-gated Ca(2+) channels (CaVs) drive synaptic transmission, neural plasticity, and cardiac contraction. Despite the importance of these events, the fundamental relationship between flux through a single CaV channel and the Ca(2+) signaling concentration within nanometers of its pore has resisted empirical determination, owing to limitations in the spatial resolution and specificity of fluorescence-based Ca(2+) measurements. Here, we exploited Ca(2+)-dependent inactivation of CaV channels as a nanometer-range Ca(2+) indicator specific to active channels. We observed an unexpected and dramatic boost in nanodomain Ca(2+) amplitude, ten-fold higher than predicted on theoretical grounds. Our results uncover a striking feature of CaV nanodomains, as diffusion-restricted environments that amplify small Ca(2+) fluxes into enormous local Ca(2+) concentrations. This Ca(2+) tuning by the physical composition of the nanodomain may represent an energy-efficient means of local amplification that maximizes information signaling capacity, while minimizing global Ca(2+) load.
Collapse
|
78
|
Abstract
Here we provide the first genome-wide in vivo analysis of the Na+/Ca2+ exchanger family in the model system Caenorhabditis elegans. We source all members of this family within the Caenorhabditis genus and reconstruct their phylogeny across humans and Drosophila melanogaster. Next, we provide a description of the expression pattern for each exchanger gene in C. elegans, revealing a wide expression in a number of tissues and cell types including sensory neurons, interneurons, motor neurons, muscle cells, and intestinal tissue. Finally, we conduct a series of behavioral and functional analyses through mutant characterization in C. elegans. From these data we demonstrate that, similar to mammalian systems, the expression of Na+/Ca2+ exchangers in C. elegans is skewed toward excitable cells, and we propose that C. elegans may be an ideal model system for the study of Na+/Ca2+ exchangers.
Collapse
|
79
|
Pilla AA. Nonthermal electromagnetic fields: From first messenger to therapeutic applications. Electromagn Biol Med 2013; 32:123-36. [DOI: 10.3109/15368378.2013.776335] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
80
|
Influence of Experimental Subarachnoid Hemorrhage on Nicotine-induced Contraction of the Rat Basilar Artery in Relation to Nicotinic Acetylcholine Receptors, Calcium, and Potassium Channels. J Stroke Cerebrovasc Dis 2013; 22:371-7. [DOI: 10.1016/j.jstrokecerebrovasdis.2011.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 09/01/2011] [Accepted: 09/27/2011] [Indexed: 11/23/2022] Open
|
81
|
Lukyanets IA, Lukyanetz EA. Modulation of calcium signalling by the endoplasmic reticulum in Carassius neurons. Biochem Biophys Res Commun 2013; 433:591-4. [DOI: 10.1016/j.bbrc.2013.03.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/14/2013] [Indexed: 11/28/2022]
|
82
|
Actin-based confinement of calcium responses during Shigella invasion. Nat Commun 2013; 4:1567. [DOI: 10.1038/ncomms2561] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 01/28/2013] [Indexed: 12/18/2022] Open
|
83
|
Hashimoto M, Enomoto M, Morales J, Kurebayashi N, Sakurai T, Hashimoto T, Nara T, Mikoshiba K. Inositol 1,4,5-trisphosphate receptor regulates replication, differentiation, infectivity and virulence of the parasitic protist Trypanosoma cruzi. Mol Microbiol 2013; 87:1133-50. [PMID: 23320762 DOI: 10.1111/mmi.12155] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2013] [Indexed: 11/26/2022]
Abstract
In animals, inositol 1,4,5-trisphosphate receptors (IP3 Rs) are ion channels that play a pivotal role in many biological processes by mediating Ca(2+) release from the endoplasmic reticulum. Here, we report the identification and characterization of a novel IP3 R in the parasitic protist, Trypanosoma cruzi, the pathogen responsible for Chagas disease. DT40 cells lacking endogenous IP3 R genes expressing T. cruzi IP3 R (TcIP3 R) exhibited IP3 -mediated Ca(2+) release from the ER, and demonstrated receptor binding to IP3 . TcIP3 R was expressed throughout the parasite life cycle but the expression level was much lower in bloodstream trypomastigotes than in intracellular amastigotes or epimastigotes. Disruption of two of the three TcIP3 R gene loci led to the death of the parasite, suggesting that IP3 R is essential for T. cruzi. Parasites expressing reduced or increased levels of TcIP3 R displayed defects in growth, transformation and infectivity, indicating that TcIP3 R is an important regulator of the parasite's life cycle. Furthermore, mice infected with T. cruzi expressing reduced levels of TcIP3 R exhibited a reduction of disease symptoms, indicating that TcIP3 R is an important virulence factor. Combined with the fact that the primary structure of TcIP3 R has low similarity to that of mammalian IP3 Rs, TcIP3 R is a promising drug target for Chagas disease.
Collapse
Affiliation(s)
- Muneaki Hashimoto
- Department of Molecular and Cellular Parasitology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Bootman MD, Rietdorf K, Collins T, Walker S, Sanderson M. Ca2+-sensitive fluorescent dyes and intracellular Ca2+ imaging. Cold Spring Harb Protoc 2013; 2013:83-99. [PMID: 23378644 DOI: 10.1101/pdb.top066050] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Imaging Ca(2+)-sensitive fluorescent indicators provides a common approach for studying Ca(2+) signals in many contexts. Fluorescent indicators are particularly useful for measuring acute Ca(2+) changes in a relatively noninvasive manner. The availability of indicators that can be targeted to specific cellular domains, coupled with variations in affinity, brightness or spectral characteristics, provides tools for exploring spatially and temporally diverse Ca(2+) signals, and moreover, multiplexing the readout of Ca(2+) with other cellular functions. This article aims to give the novice experimentalist some insight into the considerations and potential pitfalls that impinge on the use of fluorescent Ca(2+) indicators.
Collapse
Affiliation(s)
- Martin D Bootman
- Babraham Institute, Babraham, Cambridge, CB22 3AT, United Kingdom.
| | | | | | | | | |
Collapse
|
85
|
Prevarskaya N, Skryma R, Shuba Y. Targeting Ca2+transport in cancer: close reality or long perspective? Expert Opin Ther Targets 2013; 17:225-41. [DOI: 10.1517/14728222.2013.741594] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
86
|
Williams DA, Monif M, Richardson KL. Compartmentalizing genetically encoded calcium sensors. Methods Mol Biol 2013; 937:307-26. [PMID: 23007595 DOI: 10.1007/978-1-62703-086-1_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Within single cells there is a complex myriad of signaling which controls physiological process many of which are modulated, or signaled directly, by intracellular calcium ions. Understanding the exquisitely sensitive, and spatially restricted, changes in calcium has been of interest to the researcher for a number of years. Recent advances in this field have been driven by the development of genetically encoded calcium probes for detecting calcium changes within the cells specifically targeting organelles such as mitochondria, endoplasmic reticulum, and the nucleus. In this chapter the authors outline some of the available fluorescent probes, with particular emphasis on an endoplasmic reticulum targeted calcium biosensor in cell signaling studies with astrocytes, detailing experimental protocols and the interpretation of data from such probes.
Collapse
Affiliation(s)
- David A Williams
- Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.
| | | | | |
Collapse
|
87
|
Raveh A, Valitsky M, Shani L, Coorssen JR, Blank PS, Zimmerberg J, Rahamimoff R. Observations of calcium dynamics in cortical secretory vesicles. Cell Calcium 2012; 52:217-25. [PMID: 22831912 PMCID: PMC3433649 DOI: 10.1016/j.ceca.2012.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 06/06/2012] [Accepted: 06/18/2012] [Indexed: 11/15/2022]
Abstract
Calcium (Ca(2+)) dynamics were evaluated in fluorescently labeled sea urchin secretory vesicles using confocal microscopy. 71% of the vesicles examined exhibited one or more transient increases in the fluorescence signal that was damped in time. The detection of transient increases in signal was dependent upon the affinity of the fluorescence indicator; the free Ca(2+) concentration in the secretory vesicles was estimated to be in the range of ∼10 to 100 μM. Non-linear stochastic analysis revealed the presence of extra variance in the Ca(2+) dependent fluorescence signal. This noise process increased linearly with the amplitude of the Ca(2+) signal. Both the magnitude and spatial properties of this noise process were dependent upon the activity of vesicle p-type (Ca(v)2.1) Ca(2+) channels. Blocking the p-type Ca(2+) channels with ω-agatoxin decreased signal variance, and altered the spatial noise pattern within the vesicle. These fluorescence signal properties are consistent with vesicle Ca(2+) dynamics and not simply due to obvious physical properties such as gross movement artifacts or pH driven changes in Ca(2+) indicator fluorescence. The results suggest that the free Ca(2+) content of cortical secretory vesicles is dynamic; this property may modulate the exocytotic fusion process.
Collapse
Affiliation(s)
- Adi Raveh
- Department of Physiology and the Bernard Katz Minerva Centre for Cell Biophysics, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Michael Valitsky
- Department of Physiology and the Bernard Katz Minerva Centre for Cell Biophysics, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Liora Shani
- Department of Physiology and the Bernard Katz Minerva Centre for Cell Biophysics, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Jens R. Coorssen
- Department of Molecular Physiology, School of Medicine, College of Health and Science, and Molecular Medicine Research Group, University of Western Sydney, Campbelltown, Australia
| | - Paul S. Blank
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Joshua Zimmerberg
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Rami Rahamimoff
- Department of Physiology and the Bernard Katz Minerva Centre for Cell Biophysics, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
88
|
Boczek T, Lisek M, Kowalski A, Pikula S, Niewiarowska J, Wiktorska M, Zylinska L. Downregulation of PMCA2 or PMCA3 reorganizes Ca(2+) handling systems in differentiating PC12 cells. Cell Calcium 2012; 52:433-44. [PMID: 22921123 DOI: 10.1016/j.ceca.2012.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 07/30/2012] [Accepted: 08/02/2012] [Indexed: 01/02/2023]
Abstract
Changes in PMCA2 and PMCA3 expression during neuronal development are tightly linked to structural and functional modifications in Ca(2+) handling machinery. Using antisense strategy we obtained stably transfected PC12 lines with reduced level of PMCA2 or PMCA3, which were then subjected to dibutyryl-cAMP differentiation. Reduced level of neuron-specific PMCAs led to acceleration of differentiation and formation of longer neurites than in control PC12 line. Treatment with dibutyryl-cAMP was associated with retraction of growth cones and intensified formation of varicosities. In PMCA2-reduced cells development of apoptosis and DNA laddering were detected. Higher amounts of constitutive isoforms PMCA1 and PMCA4, their putative extended location to gaps left after partial removal of PMCA2 or PMCA3, together with increased SERCA may indicate the induction of compensatory mechanism in modified cells. Functional studies showed altered expression of certain types of VDCCs in PMCA-reduced cells, which correlated with their higher contribution to Ca(2+) influx. The cell response to PMCAs suppression suggests the interplay between transcription level of two opposite calcium-transporting systems i.e. voltage- and store depletion-activated channels facilitating Ca(2+) influx and calcium pumps responsible for Ca(2+) clearance, as well highlights the role of both neuron-specific PMCA isoforms in the control of PC12 cells differentiation.
Collapse
Affiliation(s)
- Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University, Lodz, Poland.
| | | | | | | | | | | | | |
Collapse
|
89
|
Rao JN, Rathor N, Zhuang R, Zou T, Liu L, Xiao L, Turner DJ, Wang JY. Polyamines regulate intestinal epithelial restitution through TRPC1-mediated Ca²+ signaling by differentially modulating STIM1 and STIM2. Am J Physiol Cell Physiol 2012; 303:C308-17. [PMID: 22592407 PMCID: PMC3423028 DOI: 10.1152/ajpcell.00120.2012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 05/14/2012] [Indexed: 11/22/2022]
Abstract
Early epithelial restitution occurs as a consequence of intestinal epithelial cell (IEC) migration after wounding, and its defective regulation is implicated in various critical pathological conditions. Polyamines stimulate intestinal epithelial restitution, but their exact mechanism remains unclear. Canonical transient receptor potential-1 (TRPC1)-mediated Ca(2+) signaling is crucial for stimulation of IEC migration after wounding, and induced translocation of stromal interaction molecule 1 (STIM1) to the plasma membrane activates TRPC1-mediated Ca(2+) influx and thus enhanced restitution. Here, we show that polyamines regulate intestinal epithelial restitution through TRPC1-mediated Ca(2+) signaling by altering the ratio of STIM1 to STIM2. Increasing cellular polyamines by ectopic overexpression of the ornithine decarboxylase (ODC) gene stimulated STIM1 but inhibited STIM2 expression, whereas depletion of cellular polyamines by inhibiting ODC activity decreased STIM1 but increased STIM2 levels. Induced STIM1/TRPC1 association by increasing polyamines enhanced Ca(2+) influx and stimulated epithelial restitution, while decreased formation of the STIM1/TRPC1 complex by polyamine depletion decreased Ca(2+) influx and repressed cell migration. Induced STIM1/STIM2 heteromers by polyamine depletion or STIM2 overexpression suppressed STIM1 membrane translocation and inhibited Ca(2+) influx and epithelial restitution. These results indicate that polyamines differentially modulate cellular STIM1 and STIM2 levels in IECs, in turn controlling TRPC1-mediated Ca(2+) signaling and influencing cell migration after wounding.
Collapse
Affiliation(s)
- Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Abstract
The versatility of Ca2+ as an intracellular messenger stems largely from the impressive, but complex, spatiotemporal organization of the Ca2+ signals. For example, the latter when initiated by IP3 (inositol 1,4,5-trisphosphate) in many cells manifest hierarchical recruitment of elementary Ca2+ release events ('blips' and then 'puffs') en route to global regenerative Ca2+ waves as the cellular IP3 concentration rises. The spacing of IP3Rs (IP3 receptors) and their regulation by Ca2+ are key determinants of these spatially organized Ca2+ signals, but neither is adequately understood. IP3Rs have been proposed to be pre-assembled into clusters, but their composition, geometry and whether clustering affects IP3R behaviour are unknown. Using patch-clamp recording from the outer nuclear envelope of DT40 cells expressing rat IP3R1 or IP3R3, we have recently shown that low concentrations of IP3 cause IP3Rs to aggregate rapidly and reversibly into small clusters of approximately four IP3Rs. At resting cytosolic Ca2+ concentrations, clustered IP3Rs open independently, but with lower open probability, shorter open duration and lesser IP3-sensitivity than lone IP3Rs. This inhibitory influence of clustering on IP3R is reversed when the [Ca2+]i (cytosolic free Ca2+ concentration) increases. The gating of clustered IP3Rs exposed to increased [Ca2+]i is coupled: they are more likely to open and close together, and their simultaneous openings are prolonged. Dynamic clustering of IP3Rs by IP3 thus exposes them to local Ca2+ rises and increases their propensity for a CICR (Ca2+-induced Ca2+ rise), thereby facilitating hierarchical recruitment of the elementary events that underlie all IP3-evoked Ca2+ signals.
Collapse
|
91
|
Groschner LN, Waldeck-Weiermair M, Malli R, Graier WF. Endothelial mitochondria--less respiration, more integration. Pflugers Arch 2012; 464:63-76. [PMID: 22382745 PMCID: PMC3387498 DOI: 10.1007/s00424-012-1085-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 02/11/2012] [Indexed: 12/21/2022]
Abstract
Lining the inner surface of the circulatory system, the vascular endothelium accomplishes a vast variety of specialized functions. Even slight alterations of these functions are implicated in the development of certain cardiovascular diseases that represent major causes of morbidity and mortality in developed countries. Endothelial mitochondria are essential to the functional integrity of the endothelial cell as they integrate a wide range of cellular processes including Ca²⁺ handling, redox signaling and apoptosis, all of which are closely interrelated. Growing evidence supports the notion that impairment of mitochondrial signaling in the endothelium is an early event and a causative factor in the development of diseases such as atherosclerosis or diabetic complications. In this review, we want to outline the significance of mitochondria in both physiology and pathology of the vascular endothelium.
Collapse
Affiliation(s)
- Lukas N. Groschner
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21/III, 8010 Graz, Austria
| | - Markus Waldeck-Weiermair
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21/III, 8010 Graz, Austria
| | - Roland Malli
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21/III, 8010 Graz, Austria
| | - Wolfgang F. Graier
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21/III, 8010 Graz, Austria
| |
Collapse
|
92
|
Pavlovic V, Djordjevic A, Cherneva E, Yancheva D, Smelcerovic A. Stimulatory effect on rat thymocytes proliferation and antimicrobial activity of two 6-(propan-2-yl)-4-methyl-morpholine-2,5-diones. Food Chem Toxicol 2012; 50:761-6. [DOI: 10.1016/j.fct.2011.11.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/03/2011] [Accepted: 11/07/2011] [Indexed: 11/24/2022]
|
93
|
Abstract
All cells use changes in intracellular calcium concentration ([Ca(2+)](i)) to regulate cell signalling events. In neurons, with their elaborate dendritic and axonal arborizations, there are clear examples of both localized and widespread Ca(2+) signals. [Ca(2+)](i) changes that are generated by Ca(2+) entry through voltage- and ligand-gated channels are the best characterized. In addition, the release of Ca(2+) from intracellular stores can result in increased [Ca(2+)](i); the signals that trigger this release have been less well-studied, in part because they are not usually associated with specific changes in membrane potential. However, recent experiments have revealed dramatic widespread Ca(2+) waves and localized spark-like events, particularly in dendrites. Here we review emerging data on the nature of these signals and their functions.
Collapse
|
94
|
Johnson JD, Bround MJ, White SA, Luciani DS. Nanospaces between endoplasmic reticulum and mitochondria as control centres of pancreatic β-cell metabolism and survival. PROTOPLASMA 2012; 249 Suppl 1:S49-S58. [PMID: 22105567 DOI: 10.1007/s00709-011-0349-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/07/2011] [Indexed: 05/31/2023]
Abstract
Nanometre-scale spaces between organelles represent focused nodes for signal transduction and the control of cellular decisions. The endoplasmic reticulum (ER) and the mitochondria form dynamic quasi-synaptic interaction nanodomains in all cell types examined, but the functional role of these junctions in cellular metabolism and cell survival remains to be fully understood. In this paper, we review recent evidence that ER Ca(2+) channels, such as the RyR and IP(3)R, can signal specifically across this nanodomain to the adjacent mitochondria to pace basal metabolism, with focus on the pancreatic β-cell. Blocking these signals in the basal state leads to a form of programmed cell death associated with reduced ATP and the induction of calpain-10 and hypoxia-inducible factors. On the other hand, the hyperactivity of this signalling domain plays a deleterious role during classical forms of apoptosis. Thus, the nanospace between ER and mitochondria represents a critical rheostat controlling both metabolism and programmed cell death. Many aspects of the mechanisms underlying this control system remain to be uncovered, and new nanotechnologies are required understand these domains at a molecular level.
Collapse
Affiliation(s)
- James D Johnson
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada.
| | | | | | | |
Collapse
|
95
|
Plasma membrane calcium pump (PMCA) isoform 4 is targeted to the apical membrane by the w-splice insert from PMCA2. Cell Calcium 2012; 51:171-8. [PMID: 22252018 DOI: 10.1016/j.ceca.2011.12.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 11/23/2011] [Accepted: 12/13/2011] [Indexed: 11/24/2022]
Abstract
Local Ca(2+) signaling requires proper targeting of the Ca(2+) signaling toolkit to specific cellular locales. Different isoforms of the plasma membrane Ca(2+) pump (PMCA) are responsible for Ca(2+) extrusion at the apical and basolateral membrane of polarized epithelial cells, but the mechanisms and signals for differential targeting of the PMCAs are not well understood. Recent work demonstrated that the alternatively spliced w-insert in PMCA2 directs this pump to the apical membrane. We now show that inserting the w-insert into the corresponding location of the PMCA4 isoform confers apical targeting to this normally basolateral pump. Mutation of a di-leucine motif in the C-tail thought to be important for basolateral targeting did not enhance apical localization of the chimeric PMCA4(2w)/b. In contrast, replacing the C-terminal Val residue by Leu to optimize the PDZ ligand site for interaction with the scaffolding protein NHERF2 enhanced the apical localization of PMCA4(2w)/b, but not of PMCA4x/b. Functional studies showed that both apical PMCA4(2w)/b and basolateral PMCA4x/b handled ATP-induced Ca(2+) signals with similar kinetics, suggesting that isoform-specific functional characteristics are retained irrespective of membrane targeting. Our results demonstrate that the alternatively spliced w-insert provides autonomous apical targeting information in the PMCA without altering its functional characteristics.
Collapse
|
96
|
Ryanodine Receptor Physiology and Its Role in Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:217-34. [DOI: 10.1007/978-94-007-2888-2_9] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
97
|
|
98
|
Tonelli FMP, Santos AK, Gomes DA, da Silva SL, Gomes KN, Ladeira LO, Resende RR. Stem cells and calcium signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:891-916. [PMID: 22453975 DOI: 10.1007/978-94-007-2888-2_40] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The increasing interest in stem cell research is linked to the promise of developing treatments for many lifethreatening, debilitating diseases, and for cell replacement therapies. However, performing these therapeutic innovations with safety will only be possible when an accurate knowledge about the molecular signals that promote the desired cell fate is reached. Among these signals are transient changes in intracellular Ca(2+) concentration [Ca(2+)](i). Acting as an intracellular messenger, Ca(2+) has a key role in cell signaling pathways in various differentiation stages of stem cells. The aim of this chapter is to present a broad overview of various moments in which Ca(2+)-mediated signaling is essential for the maintenance of stem cells and for promoting their development and differentiation, also focusing on their therapeutic potential.
Collapse
Affiliation(s)
- Fernanda M P Tonelli
- Nanomaterials Laboratory, Department of Physics, Insitute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | |
Collapse
|
99
|
Hashimoto T, Kiya M, Ohata H, Miyazaki T, Shibata K, Nobe K, Honda K. Spatiotemporal dynamics of intracellular calcium in the middle cerebral artery isolated from stroke-prone spontaneously hypertensive rats. Exp Physiol 2011; 97:265-76. [DOI: 10.1113/expphysiol.2011.061499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
100
|
Ehrlich LS, Medina GN, Carter CA. ESCRT machinery potentiates HIV-1 utilization of the PI(4,5)P(2)-PLC-IP3R-Ca(2+) signaling cascade. J Mol Biol 2011; 413:347-58. [PMID: 21875593 PMCID: PMC3193579 DOI: 10.1016/j.jmb.2011.08.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 08/05/2011] [Accepted: 08/16/2011] [Indexed: 01/09/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) release efficiency is directed by late (L) domain motifs in the viral structural precursor polyprotein Gag, which serve as links to the ESCRT (endosomal sorting complex required for transport) machinery. Linkage is normally through binding of Tsg101, an ESCRT-1 component, to the P(7)TAP motif in the p6 region of Gag. In its absence, budding is directed by binding of Alix, an ESCRT adaptor protein, to the LY(36)PX(n)L motif in Gag. We recently showed that budding requires activation of the inositol 1,4,5-triphosphate receptor (IP3R), a protein that "gates" Ca(2+) release from intracellular stores, triggers Ca(2+) cell influx and thereby functions as a major regulator of Ca(2+) signaling. In the present study, we determined whether the L domain links Gag to Ca(2+) signaling machinery. Depletion of IP3R and inactivation of phospholipase C (PLC) inhibited budding whether or not Tsg101 was bound to Gag. PLC hydrolysis of phosphatidylinositol-(4,5)-bisphosphate generates inositol (1,4,5)-triphosphate, the ligand that activates IP3R. However, with Tsg101 bound, Gag release was independent of Gq-mediated activation of PLC, and budding was readily enhanced by pharmacological stimulation of PLC. Moreover, IP3R was redistributed to the cell periphery and cytosolic Ca(2+) was elevated, events indicative of induction of Ca(2+) signaling. The results suggest that L domain function, ESCRT machinery and Ca(2+) signaling are linked events in Gag release.
Collapse
Affiliation(s)
- Lorna S. Ehrlich
- Dept. of Molecular Genetics & Microbiology, Stony Brook University, Stony Brook, NY 11794-5222, USA
| | - Gisselle N. Medina
- Dept. of Molecular Genetics & Microbiology, Stony Brook University, Stony Brook, NY 11794-5222, USA
| | - Carol A. Carter
- Dept. of Molecular Genetics & Microbiology, Stony Brook University, Stony Brook, NY 11794-5222, USA
| |
Collapse
|