1
|
Circulating mitochondrial N-formyl peptides contribute to secondary nosocomial infection in patients with septic shock. Proc Natl Acad Sci U S A 2021; 118:2018538118. [PMID: 33888581 PMCID: PMC8092466 DOI: 10.1073/pnas.2018538118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Septic shock commonly leads to multiorgan injury both directly via tissue inflammation and secondarily via hypoperfusion, but both can result in mitochondrial N-formyl peptide (mtFP) release into the circulation. However, no studies have evaluated the role of circulating mtFPs during septic shock. We found that a relatively high plasma nicotinamide adenine dinucleotide dehydrogenase subunit-6 (the most potent human mtFP) level was independently associated with the development of secondary infection in patients with septic shock and that the increased susceptibility to secondary infection is partly attributed to the suppression of polymorphonuclear leukocyte (PMN) chemotaxis by mtFP occupancy of formyl peptide receptor-1. Incorporation of these findings into therapeutic strategies may improve clinical outcomes in septic shock patients by preventing PMN chemotactic anergy. Secondary infections typically worsen outcomes of patients recovering from septic shock. Neutrophil [polymorphonuclear leukocytes (PMNs)] migration to secondarily inoculated sites may play a key role in inhibiting progression from local bacterial inoculation to secondary infection. Mitochondrial N-formyl peptide (mtFP) occupancy of formyl peptide receptor-1 (FPR1) has been shown to suppress PMN chemotaxis. Therefore, we studied the association between circulating mtFPs and the development of secondary infection in patients with septic shock. We collected clinical data and plasma samples from patients with septic shock admitted to the intensive care unit for longer than 72 h. Impacts of circulating nicotinamide adenine dinucleotide dehydrogenase subunit-6 (ND6) upon clinical outcomes were analyzed. Next, the role of ND6 in PMN chemotaxis was investigated using isolated human PMNs. Studying plasma samples from 97 patients with septic shock, we found that circulating ND6 levels at admission were independently and highly associated with the development of secondary infection (odds ratio = 30.317, 95% CI: 2.904 to 316.407, P = 0.004) and increased 90-d mortality (odds ratio = 1.572, 95% CI: 1.002 to 2.465, P = 0.049). In ex vivo experiments, ND6 pretreatment suppressed FPR1-mediated PMN chemotactic responses to bacterial peptides in the presence of multiple cytokines and chemokines, despite increased nondirectional PMN movements. Circulating mtFPs appear to contribute to the development of secondary infection and increased mortality in patients with septic shock who survive their early hyperinflammatory phase. The increased susceptibility to secondary infection is probably partly mediated by the suppression of FPR1-mediated PMN chemotaxis to secondary infected sites.
Collapse
|
2
|
Sushma, Mondal AC. Role of GPCR signaling and calcium dysregulation in Alzheimer's disease. Mol Cell Neurosci 2019; 101:103414. [PMID: 31655116 DOI: 10.1016/j.mcn.2019.103414] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), a late onset neurodegenerative disorder is characterized by the loss of memory, disordered cognitive function, caused by accumulation of amyloid-β (Aβ) peptide and neurofibrillary tangles (NFTs) in the neocortex and hippocampal brain area. Extensive research has been done on the findings of the disease etiology or pathological causes of aggregation of Aβ and hyperphosphorylation of tau protein without much promising results. Recently, calcium dysregulation has been reported to play an important role in the pathophysiology of AD. Calcium ion acts as one of the major secondary messengers, regulates many signaling pathways involved in cell survival, proliferation, differentiation, transcription and apoptosis. Calcium signaling is one of the major signaling pathways involved in the formation of memory, generation of energy and other physiological functions. It also can modulate function of many proteins upon binding. Dysregulation in calcium homeostasis leads to many physiological changes leading to neurodegenerative diseases including AD. In AD, GPCRs generate secondary messengers which regulate calcium homeostasis inside the cell and is reported to be disturbed in the pathological condition. Calcium channels and receptors present on the plasma membrane and intracellular organelle maintain calcium homeostasis through different signaling mechanisms. In this review, we have summarized the different calcium channels and receptors involved in calcium dysregulation which in turn play a critical role in the pathogenesis of AD. Understanding the role of calcium channels and GPCRs to maintain calcium homeostasis is an attempt to develop effective AD treatments.
Collapse
Affiliation(s)
- Sushma
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India.
| |
Collapse
|
3
|
Ramanadham S, Ali T, Ashley JW, Bone RN, Hancock WD, Lei X. Calcium-independent phospholipases A2 and their roles in biological processes and diseases. J Lipid Res 2015; 56:1643-68. [PMID: 26023050 DOI: 10.1194/jlr.r058701] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Indexed: 12/24/2022] Open
Abstract
Among the family of phospholipases A2 (PLA2s) are the Ca(2+)-independent PLA2s (iPLA2s) and they are designated group VI iPLA2s. In relation to secretory and cytosolic PLA2s, the iPLA2s are more recently described and details of their expression and roles in biological functions are rapidly emerging. The iPLA2s or patatin-like phospholipases (PNPLAs) are intracellular enzymes that do not require Ca(2+) for activity, and contain lipase (GXSXG) and nucleotide-binding (GXGXXG) consensus sequences. Though nine PNPLAs have been recognized, PNPLA8 (membrane-associated iPLA2γ) and PNPLA9 (cytosol-associated iPLA2β) are the most widely studied and understood. The iPLA2s manifest a variety of activities in addition to phospholipase, are ubiquitously expressed, and participate in a multitude of biological processes, including fat catabolism, cell differentiation, maintenance of mitochondrial integrity, phospholipid remodeling, cell proliferation, signal transduction, and cell death. As might be expected, increased or decreased expression of iPLA2s can have profound effects on the metabolic state, CNS function, cardiovascular performance, and cell survival; therefore, dysregulation of iPLA2s can be a critical factor in the development of many diseases. This review is aimed at providing a general framework of the current understanding of the iPLA2s and discussion of the potential mechanisms of action of the iPLA2s and related involved lipid mediators.
Collapse
Affiliation(s)
- Sasanka Ramanadham
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Tomader Ali
- Undergraduate Research Office, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jason W Ashley
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Robert N Bone
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - William D Hancock
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Xiaoyong Lei
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
4
|
De Bock M, Wang N, Decrock E, Bol M, Gadicherla AK, Culot M, Cecchelli R, Bultynck G, Leybaert L. Endothelial calcium dynamics, connexin channels and blood-brain barrier function. Prog Neurobiol 2013; 108:1-20. [PMID: 23851106 DOI: 10.1016/j.pneurobio.2013.06.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/12/2013] [Accepted: 06/18/2013] [Indexed: 01/11/2023]
Abstract
Situated between the circulation and the brain, the blood-brain barrier (BBB) protects the brain from circulating toxins while securing a specialized environment for neuro-glial signaling. BBB capillary endothelial cells exhibit low transcytotic activity and a tight, junctional network that, aided by the cytoskeleton, restricts paracellular permeability. The latter is subject of extensive research as it relates to neuropathology, edema and inflammation. A key determinant in regulating paracellular permeability is the endothelial cytoplasmic Ca(2+) concentration ([Ca(2+)]i) that affects junctional and cytoskeletal proteins. Ca(2+) signals are not one-time events restricted to a single cell but often appear as oscillatory [Ca(2+)]i changes that may propagate between cells as intercellular Ca(2+) waves. The effect of Ca(2+) oscillations/waves on BBB function is largely unknown and we here review current evidence on how [Ca(2+)]i dynamics influence BBB permeability.
Collapse
Affiliation(s)
- Marijke De Bock
- Dept. of Basic Medical Sciences, Ghent University, Ghent, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Hucho T, Suckow V, Joseph EK, Kuhn J, Schmoranzer J, Dina OA, Chen X, Karst M, Bernateck M, Levine JD, Ropers HH. Ca++/CaMKII switches nociceptor-sensitizing stimuli into desensitizing stimuli. J Neurochem 2012; 123:589-601. [PMID: 22891703 DOI: 10.1111/j.1471-4159.2012.07920.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 07/17/2012] [Accepted: 07/22/2012] [Indexed: 01/19/2023]
Abstract
Many extracellular factors sensitize nociceptors. Often they act simultaneously and/or sequentially on nociceptive neurons. We investigated if stimulation of the protein kinase C epsilon (PKCε) signaling pathway influences the signaling of a subsequent sensitizing stimulus. Central in activation of PKCs is their transient translocation to cellular membranes. We found in cultured nociceptive neurons that only a first stimulation of the PKCε signaling pathway resulted in PKCε translocation. We identified a novel inhibitory cascade to branch off upstream of PKCε, but downstream of Epac via IP3-induced calcium release. This signaling branch actively inhibited subsequent translocation and even attenuated ongoing translocation. A second 'sensitizing' stimulus was rerouted from the sensitizing to the inhibitory branch of the signaling cascade. Central for the rerouting was cytoplasmic calcium increase and CaMKII activation. Accordingly, in behavioral experiments, activation of calcium stores switched sensitizing substances into desensitizing substances in a CaMKII-dependent manner. This mechanism was also observed by in vivo C-fiber electrophysiology corroborating the peripheral location of the switch. Thus, we conclude that the net effect of signaling in nociceptors is defined by the context of the individual cell's signaling history.
Collapse
Affiliation(s)
- Tim Hucho
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Yang B, Gwozdz T, Dutko-Gwozdz J, Bolotina VM. Orai1 and Ca2+-independent phospholipase A2 are required for store-operated Icat-SOC current, Ca2+ entry, and proliferation of primary vascular smooth muscle cells. Am J Physiol Cell Physiol 2011; 302:C748-56. [PMID: 22094335 DOI: 10.1152/ajpcell.00312.2011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Store-operated Ca(2+) entry (SOCE) is important for multiple functions of vascular smooth muscle cells (SMC), which, depending of their phenotype, can resemble excitable and nonexcitable cells. Similar to nonexcitable cells, Orai1 was found to mediate Ca(2+)-selective (CRAC-like) current and SOCE in dedifferentiated cultured SMC and smooth muscle-derived cell lines. However, the role of Orai1 in cation-selective store-operated channels (cat-SOC), which are responsible for SOCE in primary SMC, remains unclear. Here we focus on primary SMC, and assess the role of Orai1 and Ca(2+)-independent phospholipase A(2) (iPLA(2)β, or PLA2G6) in activation of cat-SOC current (I(cat-SOC)), SOCE, and SMC proliferation. Using molecular, electrophysiological, imaging, and functional approaches, we demonstrate that molecular knockdown of either Orai1 or iPLA(2)β leads to similar inhibition of the whole cell cat-SOC current and SOCE in primary aortic SMC and results in significant reduction in DNA synthesis and impairment of SMC proliferation. This is the first demonstration that Orai1 and iPLA(2)β are equally important for cat-SOC, SOCE, and proliferation of primary aortic SMC.
Collapse
Affiliation(s)
- Bo Yang
- Ion Channel and Calcium Signaling Unit, Boston Univ. School of Medicine, Boston, MA 02118-2393, USA
| | | | | | | |
Collapse
|
7
|
Targeting of a common receptor shared by CXCL8 and N-Ac-PGP as a therapeutic strategy to alleviate chronic neutrophilic lung diseases. Eur J Pharmacol 2011; 667:1-5. [PMID: 21669195 DOI: 10.1016/j.ejphar.2011.05.073] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 05/09/2011] [Accepted: 05/27/2011] [Indexed: 12/21/2022]
Abstract
Persistent neutrophilia is implicated in the pathology of several chronic lung diseases and consequently targeting the signals that drive the recruitment of these cells offers a plausible therapeutic strategy. The tripeptide Pro-Gly-Pro (PGP) is a neutrophil chemoattractant derived from extracellular matrix collagen and implicated in diseases such as COPD and cystic fibrosis. It was anticipated that PGP exerts its chemoatactic activity by mimicking key sequences found within classical neutrophil chemokines, such as CXCL8, and binding their receptors, CXCR1/2. Recently, however, the role of CXCR1/2 as the receptors for PGP has been questioned. In this issue of European Journal of Pharmacology, three studies address this controversy and demonstrate CXCR1/2 to be a common receptor for CXCL8 and PGP. Accordingly, these studies demonstrate the therapeutic potential of targeting this shared receptor to simultaneously alleviate neutrophilic inflammation driven by multiple neutrophil chemoattractants.
Collapse
|
8
|
Autosomal recessive vitelliform macular dystrophy in a large cohort of vitelliform macular dystrophy patients. Retina 2011; 31:581-95. [PMID: 21273940 DOI: 10.1097/iae.0b013e318203ee60] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE To report 11 cases of autosomal recessive vitelliform macular dystrophy and to compare their molecular findings and phenotypic characteristics with those of patients with the more common and well-described dominant form of the disease. METHODS Blood samples were obtained from 435 unrelated individuals with a clinical diagnosis of vitelliform macular dystrophy and screened for mutations in the coding sequences of BEST1. Medical records and retinal photographs of selected patients were reviewed. RESULTS Nine of the 435 probands were found to have 2 plausible disease-causing variations in BEST1, while 198 individuals were found to have heterozygous variations compatible with autosomal dominant inheritance. Inheritance phase was determined in three of the recessive families. Six novel disease-causing mutations were identified among these recessive patients: Arg47Cys, IVS7-2A>G, IVS7+4G>A, Ile205del12ATCCTGCTCCAGAG, Pro274Arg, and Ile366delCAGGTGTGGC. Forty-four novel disease-causing mutations were identified among the patients with presumed autosomal dominant disease. The phenotype of patients with recessive alleles for BEST1 ranged from typical vitelliform lesions to extensive extramacular deposits. CONCLUSION The authors provide evidence that two abnormal BEST1 alleles, neither of which causes macular disease alone, can act in concert to cause early-onset vitelliform macular dystrophy.
Collapse
|
9
|
Overbeek SA, Henricks PAJ, Srienc AI, Koelink PJ, de Kruijf P, Lim HD, Smit MJ, Zaman GJR, Garssen J, Nijkamp FP, Kraneveld AD, Folkerts G. N-acetylated Proline-Glycine-Proline induced G-protein dependent chemotaxis of neutrophils is independent of CXCL8 release. Eur J Pharmacol 2011; 668:428-34. [PMID: 21458443 DOI: 10.1016/j.ejphar.2011.03.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 03/22/2011] [Accepted: 03/22/2011] [Indexed: 11/26/2022]
Abstract
Chronic inflammation in lung diseases contributes to lung tissue destruction leading to the formation of chemotactic collagen fragments such as N-acetylated Proline-Glycine-Proline (N-ac-PGP). In this study, we investigated in more detail the mechanism of action of N-ac-PGP in neutrophilic inflammation. N-ac-PGP was chemotactic for human neutrophils via pertussis toxin sensitive G protein-coupled receptors in vitro and directly activated this cell type, which led to cytosolic calcium mobilization and release of CXCL8. Furthermore, using a selective CXCR2 antagonist confirmed that N-ac-PGP-induced neutrophil chemotaxis is mediated through CXCR2 activation. To determine whether N-ac-PGP was solely responsible for the migration and activation of human neutrophils in vitro and not the released CXCL8 upon stimulation with N-ac-PGP, an antibody directed against CXCL8 was used. Performing chemotaxis and calcium influx assays in the presence of this antibody did not alter the effects of N-ac-PGP whereas effects of CXCL8 were attenuated. These experiments indicate that N-ac-PGP, in addition to the direct induction of chemotaxis, also directly activates neutrophils to release CXCL8. In vivo, this may lead in the long term to a self-maintaining situation enhanced by both N-ac-PGP and CXCL8, leading to a further increase in neutrophil infiltration and chronic inflammation.
Collapse
Affiliation(s)
- Saskia A Overbeek
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Cheng KT, Ong HL, Liu X, Ambudkar IS. Contribution of TRPC1 and Orai1 to Ca(2+) entry activated by store depletion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:435-49. [PMID: 21290310 PMCID: PMC3824974 DOI: 10.1007/978-94-007-0265-3_24] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Store-operated Ca(2+) entry (SOCE) is activated in response to depletion of the ER-Ca(2+) stores by the ER Ca(2+) sensor protein, STIM1 which oligomerizes and moves to ER/PM junctional domains where it interacts with and activates channels involved in SOCE. Two types of channel activities have been described. I(CRAC), via Ca(2+) release-activated Ca(2+) (CRAC) channel, which displays high Ca(2+) selectivity and accounts for the SOCE and cell function in T lymphocytes, mast cells, platelets, and some types of smooth muscle and endothelial cells. Orai1 has been established as the pore-forming component of CRAC channels and interaction of Orai1 with STIM1 is sufficient for generation of the CRAC channel. Store depletion also leads to activation of relatively non-selective cation currents (referred to as I(SOC)) that contribute to SOCE in several other cell types. TRPC channels, including TRPC1, TRPC3, and TRPC4, have been proposed as possible candidate channels for this Ca(2+) influx. TRPC1 is the best characterized channel in this regard and reported to contribute to endogenous SOCE in many cells types. TRPC1-mediated Ca(2+) entry and cation current in cells stimulated with agonist or thapsigargin are inhibited by low [Gd(3+)] and 10-20 μM 2APB (conditions that block SOCE). Importantly, STIM1 also associates with and gates TRPC1 via electrostatic interaction between STIM1 ((684)KK(685)) and TRPC1 ((639)DD(640)). Further, store depletion induces dynamic recruitment of a TRPC1/STIM1/Orai1 complex and knockdown of Orai1 completely abrogates TRPC1 function. Despite these findings, there has been much debate regarding the activation of TRPC1 by store depletion as well as the role of Orai1 and STIM1 in SOC channel function. This chapter summarizes recent studies and concepts regarding the contributions of Orai1 and TRPC1 to SOCE. Major unresolved questions regarding functional interaction between Orai1 and TRPC1 as well as possible mechanisms involved in the regulation of TRPC channels by store depletion will be discussed.
Collapse
Affiliation(s)
- Kwong Tai Cheng
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, NIDCR, NIH, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
11
|
Tintinger GR, Theron AJ, Steel HC, Cockeran R, Pretorius L, Anderson R. Protein kinase C promotes restoration of calcium homeostasis to platelet activating factor-stimulated human neutrophils by inhibition of phospholipase C. JOURNAL OF INFLAMMATION-LONDON 2009; 6:29. [PMID: 19874629 PMCID: PMC2777896 DOI: 10.1186/1476-9255-6-29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Accepted: 10/30/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND The role of protein kinase C (PKC) in regulating the activity of phospholipase C (PLC) in neutrophils activated with the chemoattractant, platelet-activating factor (PAF, 20 and 200 nM), was probed in the current study using the selective PKC inhibitors, GF10903X (0.5 - 1 muM) and staurosporine (400 nM). METHODS Alterations in cytosolic Ca2+, Ca2+ influx, inositol triphosphate (IP3), and leukotriene B4 production were measured using spectrofluorimetric, radiometric and competitive binding radioreceptor and immunoassay procedures, respectively. RESULTS Activation of the cells with PAF was accompanied by an abrupt increase in cytosolic Ca2+ followed by a gradual decline towards basal levels. Pretreatment of neutrophils with the PKC inhibitors significantly increased IP3 production with associated enhanced Ca2+ release from storage vesicles, prolongation of the peak cytosolic Ca2+ transients, delayed clearance and exaggerated reuptake of the cation, and markedly increased synthesis of LTB4. The alterations in Ca2+ fluxes observed with the PKC inhibitors were significantly attenuated by U73122, a PLC inhibitor, as well as by cyclic AMP-mediated upregulation of the Ca2+-resequestering endomembrane ATPase.Taken together, these observations are compatible with a mechanism whereby PKC negatively modulates the activity of PLC, with consequent suppression of IP3 production and down-regulation of Ca2+ mediated pro-inflammatory responses of PAF-activated neutrophils. CONCLUSION Although generally considered to initiate and/or amplify intracellular signalling cascades which activate and sustain the pro-inflammatory activities of neutrophils and other cell types, the findings of the current study have identified a potentially important physiological, anti-inflammatory function for PKC, at least in neutrophils.
Collapse
Affiliation(s)
- Gregory R Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, South Africa.
| | | | | | | | | | | |
Collapse
|
12
|
Zhang D, Shooshtarizadeh P, Laventie BJ, Colin DA, Chich JF, Vidic J, de Barry J, Chasserot-Golaz S, Delalande F, Van Dorsselaer A, Schneider F, Helle K, Aunis D, Prévost G, Metz-Boutigue MH. Two chromogranin a-derived peptides induce calcium entry in human neutrophils by calmodulin-regulated calcium independent phospholipase A2. PLoS One 2009; 4:e4501. [PMID: 19225567 PMCID: PMC2639705 DOI: 10.1371/journal.pone.0004501] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Accepted: 01/15/2009] [Indexed: 12/11/2022] Open
Abstract
Background Antimicrobial peptides derived from the natural processing of chromogranin A (CgA) are co-secreted with catecholamines upon stimulation of chromaffin cells. Since PMNs play a central role in innate immunity, we examine responses by PMNs following stimulation by two antimicrobial CgA-derived peptides. Methodology/Principal Findings PMNs were treated with different concentrations of CgA-derived peptides in presence of several drugs. Calcium mobilization was observed by using flow cytometry and calcium imaging experiments. Immunocytochemistry and confocal microscopy have shown the intracellular localization of the peptides. The calmodulin-binding and iPLA2 activating properties of the peptides were shown by Surface Plasmon Resonance and iPLA2 activity assays. Finally, a proteomic analysis of the material released after PMNs treatment with CgA-derived peptides was performed by using HPLC and Nano-LC MS-MS. By using flow cytometry we first observed that after 15 s, in presence of extracellular calcium, Chromofungin (CHR) or Catestatin (CAT) induce a concentration-dependent transient increase of intracellular calcium. In contrast, in absence of extra cellular calcium the peptides are unable to induce calcium depletion from the stores after 10 minutes exposure. Treatment with 2-APB (2-aminoethoxydiphenyl borate), a store operated channels (SOCs) blocker, inhibits completely the calcium entry, as shown by calcium imaging. We also showed that they activate iPLA2 as the two CaM-binding factors (W7 and CMZ) and that the two sequences can be aligned with the two CaM-binding domains reported for iPLA2. We finally analyzed by HPLC and Nano-LC MS-MS the material released by PMNs following stimulation by CHR and CAT. We characterized several factors important for inflammation and innate immunity. Conclusions/Significance For the first time, we demonstrate that CHR and CAT, penetrate into PMNs, inducing extracellular calcium entry by a CaM-regulated iPLA2 pathway. Our study highlights the role of two CgA-derived peptides in the active communication between neuroendocrine and immune systems.
Collapse
Affiliation(s)
- Dan Zhang
- INSERM U575, Physiopathologie du Système Nerveux, Strasbourg, France
- Département de Réanimation Médicale, Hôpital de Hautepierre, Strasbourg, France
- First Hospital, Chongqing University of Medical Sciences, Chongqing, China
| | | | - Benoît-Joseph Laventie
- UPRES-EA 3432, Institut de Bactériologie de la Faculté de Médecine, Université Louis Pasteur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Didier André Colin
- UPRES-EA 3432, Institut de Bactériologie de la Faculté de Médecine, Université Louis Pasteur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean-François Chich
- INSERM U575, Physiopathologie du Système Nerveux, Strasbourg, France
- INRA, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Jasmina Vidic
- INRA, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Jean de Barry
- Institut des Neurosciences Cellulaires et Intégratives, UMR 7168 CNRS-Université Louis Pasteur, Strasbourg, France
| | - Sylvette Chasserot-Golaz
- Institut des Neurosciences Cellulaires et Intégratives, UMR 7168 CNRS-Université Louis Pasteur, Strasbourg, France
| | | | - Alain Van Dorsselaer
- Laboratoire de spectrométrie de masse BioOrganique, IPHC-DSA, ULP, CNRS, UMR7178, Strasbourg, France
| | - Francis Schneider
- Département de Réanimation Médicale, Hôpital de Hautepierre, Strasbourg, France
| | - Karen Helle
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Dominique Aunis
- INSERM U575, Physiopathologie du Système Nerveux, Strasbourg, France
| | - Gilles Prévost
- UPRES-EA 3432, Institut de Bactériologie de la Faculté de Médecine, Université Louis Pasteur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | |
Collapse
|
13
|
Kim TJ, Seong J, Ouyang M, Sun J, Lu S, Hong JP, Wang N, Wang Y. Substrate rigidity regulates Ca2+ oscillation via RhoA pathway in stem cells. J Cell Physiol 2009; 218:285-93. [PMID: 18844232 PMCID: PMC2592496 DOI: 10.1002/jcp.21598] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Substrate rigidity plays crucial roles in regulating cellular functions, such as cell spreading, traction forces, and stem cell differentiation. However, it is not clear how substrate rigidity influences early cell signaling events such as calcium in living cells. Using highly sensitive Ca(2+) biosensors based on fluorescence resonance energy transfer (FRET), we investigated the molecular mechanism by which substrate rigidity affects calcium signaling in human mesenchymal stem cells (HMSCs). Spontaneous Ca(2+) oscillations were observed inside the cytoplasm and the endoplasmic reticulum (ER) using the FRET biosensors targeted at subcellular locations in cells plated on rigid dishes. Lowering the substrate stiffness to 1 kPa significantly inhibited both the magnitudes and frequencies of the cytoplasmic Ca(2+) oscillation in comparison to stiffer or rigid substrate. This Ca(2+) oscillation was shown to be dependent on ROCK, a downstream effector molecule of RhoA, but independent of actin filaments, microtubules, myosin light chain kinase, or myosin activity. Lysophosphatidic acid, which activates RhoA, also inhibited the frequency of the Ca(2+) oscillation. Consistently, either a constitutive active mutant of RhoA (RhoA-V14) or a dominant negative mutant of RhoA (RhoA-N19) inhibited the Ca(2+) oscillation. Further experiments revealed that HMSCs cultured on gels with low elastic moduli displayed low RhoA activities. Therefore, our results demonstrate that RhoA and its downstream molecule ROCK may mediate the substrate rigidity-regulated Ca(2+) oscillation, which determines the physiological functions of HMSCs.
Collapse
Affiliation(s)
- Tae-Jin Kim
- Neuroscience Program, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jihye Seong
- Neuroscience Program, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Mingxing Ouyang
- Department of Bioengineering, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jie Sun
- Department of Molecular & Integrative Physiology, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Shaoying Lu
- Department of Bioengineering, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jun Pyu Hong
- Department of Bioengineering, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ning Wang
- Departments of Mechanical Science and Engineering, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yingxiao Wang
- Neuroscience Program, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Molecular & Integrative Physiology, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
- Center for Biophysics and Computational Biology, Institute of Genomic Biology, and the Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
14
|
House SJ, Potier M, Bisaillon J, Singer HA, Trebak M. The non-excitable smooth muscle: calcium signaling and phenotypic switching during vascular disease. Pflugers Arch 2008; 456:769-85. [PMID: 18365243 PMCID: PMC2531252 DOI: 10.1007/s00424-008-0491-8] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 03/04/2008] [Indexed: 01/09/2023]
Abstract
Calcium (Ca(2+)) is a highly versatile second messenger that controls vascular smooth muscle cell (VSMC) contraction, proliferation, and migration. By means of Ca(2+) permeable channels, Ca(2+) pumps and channels conducting other ions such as potassium and chloride, VSMC keep intracellular Ca(2+) levels under tight control. In healthy quiescent contractile VSMC, two important components of the Ca(2+) signaling pathways that regulate VSMC contraction are the plasma membrane voltage-operated Ca(2+) channel of the high voltage-activated type (L-type) and the sarcoplasmic reticulum Ca(2+) release channel, Ryanodine Receptor (RyR). Injury to the vessel wall is accompanied by VSMC phenotype switch from a contractile quiescent to a proliferative motile phenotype (synthetic phenotype) and by alteration of many components of VSMC Ca(2+) signaling pathways. Specifically, this switch that culminates in a VSMC phenotype reminiscent of a non-excitable cell is characterized by loss of L-type channels expression and increased expression of the low voltage-activated (T-type) Ca(2+) channels and the canonical transient receptor potential (TRPC) channels. The expression levels of intracellular Ca(2+) release channels, pumps and Ca(2+)-activated proteins are also altered: the proliferative VSMC lose the RyR3 and the sarcoplasmic/endoplasmic reticulum Ca(2+) ATPase isoform 2a pump and reciprocally regulate isoforms of the ca(2+)/calmodulin-dependent protein kinase II. This review focuses on the changes in expression of Ca(2+) signaling proteins associated with VSMC proliferation both in vitro and in vivo. The physiological implications of the altered expression of these Ca(2+) signaling molecules, their contribution to VSMC dysfunction during vascular disease and their potential as targets for drug therapy will be discussed.
Collapse
Affiliation(s)
- Suzanne J. House
- The Center for Cardiovascular Sciences, Albany Medical College, 47, New Scotland Avenue, MC-8, Albany, NY 12208, USA
| | - Marie Potier
- The Center for Cardiovascular Sciences, Albany Medical College, 47, New Scotland Avenue, MC-8, Albany, NY 12208, USA
| | - Jonathan Bisaillon
- The Center for Cardiovascular Sciences, Albany Medical College, 47, New Scotland Avenue, MC-8, Albany, NY 12208, USA
| | - Harold A. Singer
- The Center for Cardiovascular Sciences, Albany Medical College, 47, New Scotland Avenue, MC-8, Albany, NY 12208, USA
| | - Mohamed Trebak
- The Center for Cardiovascular Sciences, Albany Medical College, 47, New Scotland Avenue, MC-8, Albany, NY 12208, USA
| |
Collapse
|
15
|
Chiu WT, Tang MJ, Jao HC, Shen MR. Soft substrate up-regulates the interaction of STIM1 with store-operated Ca2+ channels that lead to normal epithelial cell apoptosis. Mol Biol Cell 2008; 19:2220-30. [PMID: 18337467 PMCID: PMC2366837 DOI: 10.1091/mbc.e07-11-1170] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 02/12/2008] [Accepted: 02/29/2008] [Indexed: 11/11/2022] Open
Abstract
We have demonstrated that soft substrate induced apoptosis in polarized cells, but not in transformed cells by disturbance of Ca(2+) homeostasis. This study aims to further investigate the regulatory mechanisms underlying the disruption of Ca(2+)-signaling integrity in soft substrate-induced epithelial apoptosis. Soft substrate up-regulated the store-operated Ca(2+) (SOC) entry across the plasma membrane of normal cervical epithelial cells, which resulted in increased cytosolic Ca(2+) levels. Concomitantly, soft substrate induced the aggregation and translocation of stromal interacting molecule 1 (STIM1) toward the cell periphery to colocalize with Orai1, an essential pore subunit of SOC channel, detected by fluorescence resonance energy transfer approach and confocal image analyses. The disturbed Ca(2+) homeostasis resulted in the activation of mu-calpain, which cleaved alpha-spectrin, induced actin disorganization, and caused apoptosis. In contrast, soft substrate did not disturb Ca(2+) homeostasis or induce apoptosis in cervical cancer cells. Chelating extracellular Ca(2+) by EGTA and down-regulated SOC entry by small interfering RNA targeting STIM1 or inhibitors targeting Ca(2+)-binding site of calpain significantly inhibited soft substrate-induced activation of mu-calpain and epithelial cell apoptosis. Thus, soft substrate up-regulates the interaction of STIM1 with SOC channels, which results in the activation of mu-calpain and subsequently induces normal epithelial cell apoptosis.
Collapse
Affiliation(s)
| | - Ming-Jer Tang
- Department of Physiology
- Center for Gene Regulation and Signal Transduction Research, and
| | | | - Meng-Ru Shen
- Center for Gene Regulation and Signal Transduction Research, and
- Departments of Pharmacology and
- Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
16
|
Choi JH, Jo H, Hong JH, Lee SI, Shin DM. Alteration of expression of Ca2+ signaling proteins and adaptation of Ca2+ signaling in SERCA2+/- mouse parotid acini. Yonsei Med J 2008; 49:311-21. [PMID: 18452270 PMCID: PMC2615323 DOI: 10.3349/ymj.2008.49.2.311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
PURPOSE The sarco/endoplasmic reticulum Ca2+-ATPase (SERCA), encoded by ATP2A2, is an essential component for G-protein coupled receptor (GPCR)-dependent Ca2+ signaling. However, whether the changes in Ca2+ signaling and Ca2+ signaling proteins in parotid acinar cells are affected by a partial loss of SERCA2 are not known. MATERIALS AND METHODS In SERCA2+/- mouse parotid gland acinar cells, Ca2+ signaling, expression levels of Ca2+ signaling proteins, and amylase secretion were investigated. RESULTS SERCA2+/- mice showed decreased SERCA2 expression and an upregulation of the plasma membrane Ca2+ ATPase. A partial loss of SERCA2 changed the expression level of 1, 4, 5-tris-inositolphosphate receptors (IP3Rs), but the localization and activities of IP3Rs were not altered. In SERCA2+/- mice, muscarinic stimulation resulted in greater amylase release, and the expression of synaptotagmin was increased compared to wild type mice. CONCLUSION These results suggest that a partial loss of SERCA2 affects the expression and activity of Ca2+ signaling proteins in the parotid gland acini, however, overall Ca2+ signaling is unchanged.
Collapse
Affiliation(s)
- Jong-Hoon Choi
- Department of Oral Medicine, Yonsei University College of Dentistry, 250 Seongsanno, Seodaemun-gu, Seoul 120-752, Korea
| | | | | | | | | |
Collapse
|
17
|
Kuehn HS, Beaven MA, Ma HT, Kim MS, Metcalfe DD, Gilfillan AM. Synergistic activation of phospholipases Cgamma and Cbeta: a novel mechanism for PI3K-independent enhancement of FcepsilonRI-induced mast cell mediator release. Cell Signal 2008; 20:625-36. [PMID: 18207701 PMCID: PMC2692074 DOI: 10.1016/j.cellsig.2007.11.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 11/19/2007] [Accepted: 11/20/2007] [Indexed: 10/24/2022]
Abstract
Antigen/IgE-mediated mast cell activation via FcvarepsilonRI can be markedly enhanced by the activation of other receptors expressed on mast cells and these receptors may thus contribute to the allergic response in vivo. One such receptor family is the G protein-coupled receptors (GPCRs). Although the signaling cascade linking FcvarepsilonRI aggregation to mast cell activation has been extensively investigated, the mechanisms by which GPCRs amplify this response are relatively unknown. To investigate this, we utilized prostaglandin (PG)E2 based on initial studies demonstrating its greater ability to augment antigen-mediated degranulation in mouse mast cells than other GPCR agonists examined. This enhancement, and the ability of PGE2 to amplify antigen-induced calcium mobilization, was independent of phosphoinositide 3-kinase but was linked to a pertussis toxin-sensitive synergistic translocation to the membrane of phospholipase (PL)Cgamma and PLCbeta and to an enhancement of PLCgamma phosphorylation. This "trans-synergistic" activation of PLCbeta and gamma, in turn, enhanced production of inositol 1,4,5-trisphosphate, store-operated calcium entry, and activation of protein kinase C (PKC) (alpha and beta). These responses were critical for the promotion of degranulation. This is the first report of synergistic activation between PLCgamma and PLCbeta that permits reinforcement of signals for degranulation in mast cells.
Collapse
Affiliation(s)
- Hye Sun Kuehn
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive MSC 1881, Bethesda, MD 20892-1881, USA
| | - Michael A. Beaven
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong-Tao Ma
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mi-Sun Kim
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive MSC 1881, Bethesda, MD 20892-1881, USA
| | - Dean D. Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive MSC 1881, Bethesda, MD 20892-1881, USA
| | - Alasdair M. Gilfillan
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive MSC 1881, Bethesda, MD 20892-1881, USA
| |
Collapse
|
18
|
Cheng KT, Liu X, Ong HL, Ambudkar IS. Functional requirement for Orai1 in store-operated TRPC1-STIM1 channels. J Biol Chem 2008; 283:12935-40. [PMID: 18326500 DOI: 10.1074/jbc.c800008200] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Orai1 and TRPC1 have been proposed as core components of store-operated calcium release-activated calcium (CRAC) and store-operated calcium (SOC) channels, respectively. STIM1, a Ca(2+) sensor protein in the endoplasmic reticulum, interacts with and mediates store-dependent regulation of both channels. We have previously reported that dynamic association of Orai1, TRPC1, and STIM1 is involved in activation of store-operated Ca(2+) entry (SOCE) in salivary gland cells. In this study, we have assessed the molecular basis of TRPC1-SOC channels in HEK293 cells. We report that TRPC1+STIM1-dependent SOCE requires functional Orai1. Thapsigargin stimulation of cells expressing Orai1+STIM1 increased Ca(2+) entry and activated typical I(CRAC) current. STIM1 alone did not affect SOCE, whereas expression of Orai1 induced a decrease. Expression of TRPC1 induced a small increase in SOCE, which was greatly enhanced by co-expression of STIM1. Thapsigargin stimulation of cells expressing TRPC1+STIM1 activated a non-selective cation current, I(SOC), that was blocked by 1 microm Gd(3+) and 2-APB. Knockdown of Orai1 decreased endogenous SOCE as well as SOCE with TRPC1 alone. siOrai1 also significantly reduced SOCE and I(SOC) in cells expressing TRPC1+STIM1. Expression of R91WOrai1 or E106QOrai1 induced similar attenuation of TRPC1+STIM1-dependent SOCE and I(SOC), whereas expression of Orai1 with TRPC1+STIM1 resulted in SOCE that was larger than that with Orai1+STIM1 or TRPC1+STIM1 but not additive. Additionally, Orai1, E106QOrai1, and R91WOrai1 co-immunoprecipitated with similar levels of TRPC1 and STIM1 from HEK293 cells, and endogenous TRPC1, STIM1, and Orai1 were co-immunoprecipitated from salivary glands. Together, these data demonstrate a functional requirement for Orai1 in TRPC1+STIM1-dependent SOCE.
Collapse
Affiliation(s)
- Kwong Tai Cheng
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
19
|
Mishra RS, Carnevale KA, Cathcart MK. iPLA2beta: front and center in human monocyte chemotaxis to MCP-1. ACTA ACUST UNITED AC 2008; 205:347-59. [PMID: 18208975 PMCID: PMC2271028 DOI: 10.1084/jem.20071243] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) directs migration of blood monocytes to inflamed tissues. Despite the central role of chemotaxis in immune responses, the regulation of chemotaxis by signal transduction pathways and their in vivo significance remain to be thoroughly deciphered. In this study, we examined the intracellular location and functions of two recently identified regulators of chemotaxis, Ca2+-independent phospholipase (iPLA2β) and cytosolic phospholipase (cPLA2α), and substantiate their in vivo importance. These enzymes are cytoplasmic in unstimulated monocytes. Upon MCP-1 stimulation, iPLA2β is recruited to the membrane-enriched pseudopod. In contrast, cPLA2α is recruited to the endoplasmic reticulum. Although iPLA2β or cPLA2α antisense oligodeoxyribonucleotide (ODN)–treated monocytes display reduced speed, iPLA2β also regulates directionality and actin polymerization. iPLA2β or cPLA2α antisense ODN–treated adoptively transferred mouse monocytes display a profound defect in migration to the peritoneum in vivo. These converging observations reveal that iPLA2β and cPLA2α regulate monocyte migration from different intracellular locations, with iPLA2β acting as a critical regulator of the cellular compass, and identify them as potential targets for antiinflammatory strategies.
Collapse
Affiliation(s)
- Ravi S Mishra
- Department of Cell Biology, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
20
|
Borges S, Lindstrom S, Walters C, Warrier A, Wilson M. Discrete influx events refill depleted Ca2+ stores in a chick retinal neuron. J Physiol 2007; 586:605-26. [PMID: 18033816 DOI: 10.1113/jphysiol.2007.143339] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The depletion of ER Ca2+ stores, following the release of Ca2+ during intracellular signalling, triggers the Ca2+ entry across the plasma membrane known as store-operated calcium entry (SOCE). We show here that brief, local [Ca2+]i increases (motes) in the thin dendrites of cultured retinal amacrine cells derived from chick embryos represent the Ca2+ entry events of SOCE and are initiated by sphingosine-1-phosphate (S1P), a sphingolipid with multiple cellular signalling roles. Externally applied S1P elicits motes but not through a G protein-coupled membrane receptor. The endogenous precursor to S1P, sphingosine, also elicits motes but its action is suppressed by dimethylsphingosine (DMS), an inhibitor of sphingosine phosphorylation. DMS also suppresses motes induced by store depletion and retards the refilling of depleted stores. These effects are reversed by exogenously applied S1P. In these neurons formation of S1P is a step in the SOCE pathway that promotes Ca2+ entry in the form of motes.
Collapse
|
21
|
Liu X, Cheng KT, Bandyopadhyay BC, Pani B, Dietrich A, Paria BC, Swaim WD, Beech D, Yildrim E, Singh BB, Birnbaumer L, Ambudkar IS. Attenuation of store-operated Ca2+ current impairs salivary gland fluid secretion in TRPC1(-/-) mice. Proc Natl Acad Sci U S A 2007; 104:17542-7. [PMID: 17956991 PMCID: PMC2077292 DOI: 10.1073/pnas.0701254104] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Indexed: 12/15/2022] Open
Abstract
Agonist-induced Ca(2+) entry via store-operated Ca(2+) (SOC) channels is suggested to regulate a wide variety of cellular functions, including salivary gland fluid secretion. However, the molecular components of these channels and their physiological function(s) are largely unknown. Here we report that attenuation of SOC current underlies salivary gland dysfunction in mice lacking transient receptor potential 1 (TRPC1). Neurotransmitter-regulated salivary gland fluid secretion in TRPC1-deficient TRPC1(-/-) mice was severely decreased (by 70%). Further, agonist- and thapsigargin-stimulated SOC channel activity was significantly reduced in salivary gland acinar cells isolated from TRPC1(-/-) mice. Deletion of TRPC1 also eliminated sustained Ca(2+)-dependent potassium channel activity, which depends on Ca(2+) entry and is required for fluid secretion. Expression of key proteins involved in fluid secretion and Ca(2+) signaling, including STIM1 and other TRPC channels, was not altered. Together, these data demonstrate that reduced SOC entry accounts for the severe loss of salivary gland fluid secretion in TRPC1(-/-) mice. Thus, TRPC1 is a critical component of the SOC channel in salivary gland acinar cells and is essential for neurotransmitter-regulation of fluid secretion.
Collapse
Affiliation(s)
- Xibao Liu
- *Secretory Physiology Section, Gene Therapy and Therapeutic Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Kwong Tai Cheng
- *Secretory Physiology Section, Gene Therapy and Therapeutic Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Bidhan C. Bandyopadhyay
- *Secretory Physiology Section, Gene Therapy and Therapeutic Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202
| | - Biswaranjan Pani
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202
| | - Alexander Dietrich
- Institute for Pharmacology and Toxicology, Philipps University Marburg, 35043 Marburg, Germany
| | - Biman C. Paria
- *Secretory Physiology Section, Gene Therapy and Therapeutic Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - William D. Swaim
- *Secretory Physiology Section, Gene Therapy and Therapeutic Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - David Beech
- Institute of Membrane and Systems Biology, University of Leeds, Leeds LS2 9JT, United Kingdom; and
| | - Eda Yildrim
- Laboratory of Signal Transduction, National Institute on Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Brij B. Singh
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202
| | - Lutz Birnbaumer
- Laboratory of Signal Transduction, National Institute on Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Indu S. Ambudkar
- *Secretory Physiology Section, Gene Therapy and Therapeutic Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
22
|
Ambudkar IS, Ong HL. Organization and function of TRPC channelosomes. Pflugers Arch 2007; 455:187-200. [PMID: 17486362 DOI: 10.1007/s00424-007-0252-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2007] [Accepted: 03/10/2007] [Indexed: 12/20/2022]
Abstract
TRPC proteins constitute a family of conserved Ca2+-permeable cation channels which are activated in response to agonist-stimulated PIP2 hydrolysis. These channels were initially proposed to be components of the store-operated calcium entry channel (SOC). Subsequent studies have provided substantial evidence that some TRPCs contribute to SOC activity. TRPC proteins have also been shown to form agonist-stimulated calcium entry channels that are not store-operated but are likely regulated by PIP2 or diacylglycerol. Further, and consistent with the presently available data, selective homomeric or heteromeric interactions between TRPC monomers generate distinct agonist-stimulated cation permeable channels. We suggest that interaction between TRPC monomers, as well as the association of these channels with accessory proteins, determines their mode of regulation as well as their cellular localization and function. Currently identified accessory proteins include key Ca2+ signaling proteins as well as proteins involved in vesicle trafficking, cytoskeletal interactions, and scaffolding. Studies reported until now demonstrate that TRPC proteins are segregated into specific Ca2+ signaling complexes which can generate spatially and temporally controlled [Ca2+]i signals. Thus, the functional organization of TRPC channelosomes dictates not only their regulation by extracellular stimuli but also serves as a platform to coordinate specific downstream cellular functions that are regulated as a consequence of Ca2+ entry. This review will focus on the accessory proteins of TRPC channels and discuss the functional implications of TRPC channelosomes and their assembly in microdomains.
Collapse
Affiliation(s)
- Indu S Ambudkar
- Secretory Physiology Section, Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
23
|
Ong HL, Cheng KT, Liu X, Bandyopadhyay BC, Paria BC, Soboloff J, Pani B, Gwack Y, Srikanth S, Singh BB, Gill D, Ambudkar IS. Dynamic assembly of TRPC1-STIM1-Orai1 ternary complex is involved in store-operated calcium influx. Evidence for similarities in store-operated and calcium release-activated calcium channel components. J Biol Chem 2007; 282:9105-16. [PMID: 17224452 PMCID: PMC3309402 DOI: 10.1074/jbc.m608942200] [Citation(s) in RCA: 315] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Store-operated calcium entry (SOCE) is a ubiquitous mechanism that is mediated by distinct SOC channels, ranging from the highly selective calcium release-activated Ca2+ (CRAC) channel in rat basophilic leukemia and other hematopoietic cells to relatively Ca2+-selective or non-selective SOC channels in other cells. Although the exact composition of these channels is not yet established, TRPC1 contributes to SOC channels and regulation of physiological function of a variety of cell types. Recently, Orai1 and STIM1 have been suggested to be sufficient for generating CRAC channels. Here we show that Orai1 and STIM1 are also required for TRPC1-SOC channels. Knockdown of TRPC1, Orai1, or STIM1 attenuated, whereas overexpression of TRPC1, but not Orai1 or STIM1, induced an increase in SOC entry and I(SOC) in human salivary gland cells. All three proteins were co-localized in the plasma membrane region of cells, and thapsigargin increased co-immunoprecipitation of TRPC1 with STIM1, and Orai1 in human salivary gland cells as well as dispersed mouse submandibular gland cells. In aggregate, the data presented here reveal that all three proteins are essential for generation of I(SOC) in these cells and that dynamic assembly of TRPC1-STIM1-Orai1 ternary complex is involved in activation of SOC channel in response to internal Ca2+ store depletion. Thus, these data suggest a common molecular basis for SOC and CRAC channels.
Collapse
Affiliation(s)
- Hwei Ling Ong
- Secretory Physiology Section, Gene Therapy and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Kwong Tai Cheng
- Secretory Physiology Section, Gene Therapy and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Xibao Liu
- Secretory Physiology Section, Gene Therapy and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Bidhan C. Bandyopadhyay
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota 58203
| | - Biman C. Paria
- Secretory Physiology Section, Gene Therapy and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Jonathan Soboloff
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Biswaranjan Pani
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota 58203
| | - Yousang Gwack
- Department of Pathology, Harvard Medical School and the CBR Institute of Biomedical Research, Boston, Massachusetts 02115
| | - Sonal Srikanth
- Department of Pathology, Harvard Medical School and the CBR Institute of Biomedical Research, Boston, Massachusetts 02115
| | - Brij B. Singh
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota 58203
| | - Donald Gill
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Indu S. Ambudkar
- Secretory Physiology Section, Gene Therapy and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
24
|
Godin N, Rousseau E. TRPC6 silencing in primary airway smooth muscle cells inhibits protein expression without affecting OAG-induced calcium entry. Mol Cell Biochem 2006; 296:193-201. [PMID: 16977347 DOI: 10.1007/s11010-006-9309-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2006] [Accepted: 08/11/2006] [Indexed: 10/24/2022]
Abstract
TRPC proteins have been described as non-selective cation channels and are thought to be involved in the regulation of Ca(2+) movement in various cells, including airway smooth muscle (ASM) cells. In order to study the role of these channels in ASM cells, transfection of a small interfering RNA (siRNA) designed against the TRPC6 channel was performed in guinea pig primary ASM cells. This specific siRNA was complexed with the new X-TremeGene (X-TG) chemical transfection reagent, whose efficiency and low cytotoxicity were determined by the use of a non-silencing rhodamine-tagged siRNA. It was found that more than 95% of cells were transfected by an optimized protocol. Verification of TRPC6 transcript down-regulation was determined by RT-PCR while Western blot analysis attested to lower protein content in the microsomal fraction. Micro-spectrofluorimetry measurements of control and siRNA-treated cells revealed that lower TRPC6 expression did not affect OAG-induced intracellular Ca(2+) movement. Thus, TRPC6 channels cannot be defined as simple Ca(2+) transporters but more likely as protein complexes supporting monovalent cation conductance in ASM cells. These conductances would in turn facilitate membrane depolarization of high input resistance cells, Ca(2+) channel activation and tone increase. In conclusion, this study defines a valuable model of RNA interference study in primary cultures of ASM cells, eventually allowing for silencing of other target proteins for which no pharmacological modulators are currently available.
Collapse
Affiliation(s)
- Nicolas Godin
- Le Bilarium, Department of Physiology and Biophysics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th avenue north, J1H 5N4, Sherbrooke, QC, Canada
| | | |
Collapse
|
25
|
Means S, Smith AJ, Shepherd J, Shadid J, Fowler J, Wojcikiewicz RJH, Mazel T, Smith GD, Wilson BS. Reaction diffusion modeling of calcium dynamics with realistic ER geometry. Biophys J 2006; 91:537-57. [PMID: 16617072 PMCID: PMC1483115 DOI: 10.1529/biophysj.105.075036] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Accepted: 03/15/2006] [Indexed: 11/18/2022] Open
Abstract
We describe a finite-element model of mast cell calcium dynamics that incorporates the endoplasmic reticulum's complex geometry. The model is built upon a three-dimensional reconstruction of the endoplasmic reticulum (ER) from an electron tomographic tilt series. Tetrahedral meshes provide volumetric representations of the ER lumen, ER membrane, cytoplasm, and plasma membrane. The reaction-diffusion model simultaneously tracks changes in cytoplasmic and ER intraluminal calcium concentrations and includes luminal and cytoplasmic protein buffers. Transport fluxes via PMCA, SERCA, ER leakage, and Type II IP3 receptors are also represented. Unique features of the model include stochastic behavior of IP3 receptor calcium channels and comparisons of channel open times when diffusely distributed or aggregated in clusters on the ER surface. Simulations show that IP3R channels in close proximity modulate activity of their neighbors through local Ca2+ feedback effects. Cytoplasmic calcium levels rise higher, and ER luminal calcium concentrations drop lower, after IP3-mediated release from receptors in the diffuse configuration. Simulation results also suggest that the buffering capacity of the ER, and not restricted diffusion, is the predominant factor influencing average luminal calcium concentrations.
Collapse
Affiliation(s)
- Shawn Means
- Sandia National Laboratory, Albuquerque, New Mexico, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Brueggemann LI, Markun DR, Henderson KK, Cribbs LL, Byron KL. Pharmacological and electrophysiological characterization of store-operated currents and capacitative Ca(2+) entry in vascular smooth muscle cells. J Pharmacol Exp Ther 2006; 317:488-99. [PMID: 16415091 DOI: 10.1124/jpet.105.095067] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Capacitative Ca(2+) entry (CCE) in vascular smooth muscle cells contributes to vasoconstrictor and mitogenic effects of vasoactive hormones. In A7r5 rat aortic smooth muscle cells, measurements of cytosolic free Ca(2+) concentration ([Ca(2+)](i)) have demonstrated that depletion of intracellular Ca(2+) stores activates CCE. However, there is disagreement in published studies regarding the regulation of this mechanism by the vasoconstrictor hormone [Arg(8)]-vasopressin (AVP). We have employed electrophysiological methods to characterize the membrane currents activated by store depletion [store-operated current (I(SOC))]. Because of different recording conditions, it has not been previously determined whether I(SOC) corresponds to CCE measured using fura-2; nor has the channel protein responsible for CCE been identified. In the present study, the pharmacological characteristics of I(SOC), including its sensitivity to blockade by 2-aminoethoxydiphenylborane, diethylstilbestrol, or micromolar Gd(3+), were found to parallel the effects of these drugs on thapsigargin- or AVP-activated CCE measured under identical external ionic conditions using fura-2. Thapsigargin-stimulated I(SOC) was also measured in freshly isolated rat mesenteric artery smooth muscle cells (MASMC). Members of the transient receptor potential (TRP) family of nonselective cation channels, TRPC1, TRPC4, and TRPC6, were detected by reverse transcription-polymerase chain reaction and Western blot in both A7r5 cells and MASMC. TRPC1 expression was reduced in a stable A7r5 cell line expressing a small interfering RNA (siRNA) or by infection of A7r5 cells with an adenovirus expressing a TRPC1 antisense nucleotide sequence. Thapsigargin-stimulated I(SOC) was reduced in both the TRPC1 siRNA- and TRPC1 antisense-expressing cells, suggesting that the TRPC1 channel contributes to the I(SOC)/CCE pathway.
Collapse
Affiliation(s)
- Lioubov I Brueggemann
- Department of Pharmacology and Experimental Therapeutics, Loyola University Medical Center, 2160 South First Avenue, Maywood, IL 60153, USA
| | | | | | | | | |
Collapse
|
27
|
Ambudkar IS. Ca2+ signaling microdomains:platforms for the assembly and regulation of TRPC channels. Trends Pharmacol Sci 2005; 27:25-32. [PMID: 16337693 DOI: 10.1016/j.tips.2005.11.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Revised: 10/05/2005] [Accepted: 11/21/2005] [Indexed: 12/23/2022]
Abstract
The transient receptor potential canonical family (TRPC1-TRPC7) of ion channel proteins, which are activated in response to agonist-stimulated phosphatidylinositol (4,5)-bisphosphate [PtdIns(4,5)P(2)] hydrolysis, are proposed components of the elusive store-operated Ca(2+) (SOC) channel. TRPC channels display distinct properties and interact to form homomeric or heteromeric channels that differ in their function and regulation. Although the exact function of TRPC channels and how they are regulated has not been established, increasing data suggest that they are localized and regulated within Ca(2+) signaling microdomains. TRPC channels contribute to store-operated and store-independent Ca(2+) entry mechanisms, both of which are activated by agonist-stimulated PtdIns(4,5)P(2) hydrolysis. Elucidation of how cells achieve specificity and precise temporal and spatial coordination of channel activation is crucial for understanding the molecular basis of agonist-mediated stimulation of Ca(2+) entry and identifying downstream physiological functions. This review will address the assembly and localization of TRPC channels and how these processes impact their function.
Collapse
Affiliation(s)
- Indu S Ambudkar
- Secretory Physiology Section, GTTB, National Institute of Dental and Craniofacial Research/NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Bolotina VM, Csutora P. CIF and other mysteries of the store-operated Ca2+-entry pathway. Trends Biochem Sci 2005; 30:378-87. [PMID: 15951181 DOI: 10.1016/j.tibs.2005.05.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2005] [Revised: 05/06/2005] [Accepted: 05/26/2005] [Indexed: 11/25/2022]
Abstract
The molecular mechanism of the store-operated Ca2+-entry (SOCE) pathway remains one of the most intriguing and long lasting mysteries of Ca2+ signaling. The elusive calcium influx factor (CIF) that is produced upon depletion of Ca2+ stores has attracted growing attention, triggered by new discoveries that filled the gap in the chain of reactions leading to activation of store-operated channels and Ca2+ entry. Ca2+-independent phospholipase A2 emerged as a target of CIF, and a major determinant of the SOCE mechanism. Here, we present our viewpoint on CIF and conformational-coupling models of SOCE from a historical perspective, trying to resolve some of the problem areas, and summarizing our present knowledge on how depletion of intracellular Ca2+ stores signals to plasma membrane channels to open and provide Ca2+ influx that is required for many important physiological functions.
Collapse
Affiliation(s)
- Victoria M Bolotina
- Ion Channel and Calcium Unit, Boston University School of Medicine, Boston, MA 02118, USA.
| | | |
Collapse
|
29
|
Xie Q, Zhang Y, Sun XC, Zhai C, Bonanno JA. Expression and functional evaluation of transient receptor potential channel 4 in bovine corneal endothelial cells. Exp Eye Res 2005; 81:5-14. [PMID: 15978249 PMCID: PMC4110920 DOI: 10.1016/j.exer.2005.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2004] [Revised: 01/06/2005] [Accepted: 01/07/2005] [Indexed: 10/25/2022]
Abstract
We previously found that activation of purinergic receptors mobilizes Ca2+ and enhances bicarbonate transport in bovine corneal endothelial cells (BCEC). Since transient receptor potential channel 4 (TRPC) has been reported to be a candidate for capacitative calcium entry (CCE) and receptor operated calcium entry (ROC), we examined the expression of TRPC4 and evaluated the potential involvement of TRPC4 in CCE or ROC in BCEC. The C-terminus of TRPC4 was fused into the glutathione S-transferase (GST) expression vector. The fusion protein GST-TRPC4c was induced in bacteria and purified by affinity chromatography. An antibody was raised in rabbit by using the purified GST-TRPC4c antigen. In Western blotting, the TRPC4 antibody recognized the fusion protein while the pre-immune IgG did not. The TRPC4 antibody recognized a band at around 80 kD for membrane proteins from both the fresh and cultured BCEC. The pre-immune IgG could not detect bands at the same size. Incubation with the TRPC4c antigen abolished the 80 kD band. Immunofluorescence using the TRPC4 antibody stained both fresh and cultured BCEC, while pre-immune IgG did not. RNAi knocked down the expression of TRPC4 in cultured BCEC. Ca2+ entry induced by the purinergic receptor agonist ATP, was increased in TRPC4-siRNA transfected cells compared with the scrambled siRNA control, while Ca2+ entry induced by store depletion through blocking the endoplasmic reticulum Ca2+ pump, did not differ between the siRNA and scrambled siRNA-treated cells. Taken together, these results show that TRPC4 protein is expressed in the bovine corneal endothelial cells and may be a negative regulator in ROC stimulated by purinergic activation, but not by store depletion itself.
Collapse
Affiliation(s)
| | | | | | | | - Joseph A. Bonanno
- Corresponding author. Dr Joseph A. Bonanno, School of Optometry, Indiana University, 800 E Atwater Avenue, Bloomington, IN 47405-3680, USA. (J.A. Bonanno)
| |
Collapse
|
30
|
Bouron A, Altafaj X, Boisseau S, De Waard M. A store-operated Ca2+ influx activated in response to the depletion of thapsigargin-sensitive Ca2+ stores is developmentally regulated in embryonic cortical neurons from mice. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 159:64-71. [PMID: 16099516 DOI: 10.1016/j.devbrainres.2005.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 07/06/2005] [Accepted: 07/10/2005] [Indexed: 11/29/2022]
Abstract
Store-operated channels (SOCs) are recruited in response to the release of Ca2+ from intracellular stores. They allow a voltage-independent entry of Ca2+ into the cytoplasm also termed capacitative Ca2+ entry (CCE). In neurons, the functional significance of this Ca2+ route remains elusive. Several reports indicate that SOCs could be developmentally regulated. We verified the presence of a CCE in freshly dissociated cortical cells from E13, E14, E16, E18 fetuses and from 1-day-old mice. Intracellular Ca2+ stores were depleted by means of the SERCA pump inhibitor thapsigargin. At E13, the release of Ca2+ from thapsigargin-sensitive compartments gave rise to an entry of Ca2+ in a minority of cells. This Ca2+ route, insensitive to voltage-gated Ca2+ channel antagonists like Cd2+ and Ni2+, was blocked by the SOC inhibitor SKF-96365. After E13 and on E13 cells kept in culture, there is a marked increase in the percentage of cells with functional SOCs. The lanthanide La3+ fully inhibited the CCE from neonatal mice whereas it weakly blocked the thapsigargin-dependent Ca2+ entry at E13. This suggests that the subunit composition of the cortical SOCs is developmentally regulated with La3+-insensitive channels being expressed in the embryonic cortex whereas La3+-sensitive SOCs are found at birth. Our data argue for the presence of SOCs in embryonic cortical neurons. Their expression and pharmacological properties are developmentally regulated.
Collapse
Affiliation(s)
- Alexandre Bouron
- Laboratoire Canaux Calciques Fonctions et Pathologies, Inserm U607, DRDC/CEA, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France.
| | | | | | | |
Collapse
|
31
|
van der Stelt M, Trevisani M, Vellani V, De Petrocellis L, Schiano Moriello A, Campi B, McNaughton P, Geppetti P, Di Marzo V. Anandamide acts as an intracellular messenger amplifying Ca2+ influx via TRPV1 channels. EMBO J 2005; 24:3026-37. [PMID: 16107881 PMCID: PMC1201361 DOI: 10.1038/sj.emboj.7600784] [Citation(s) in RCA: 189] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2005] [Accepted: 07/21/2005] [Indexed: 11/09/2022] Open
Abstract
The endocannabinoid anandamide is able to interact with the transient receptor potential vanilloid 1 (TRPV1) channels at a molecular level. As yet, endogenously produced anandamide has not been shown to activate TRPV1, but this is of importance to understand the physiological function of this interaction. Here, we show that intracellular Ca2+ mobilization via the purinergic receptor agonist ATP, the muscarinic receptor agonist carbachol or the Ca(2+)-ATPase inhibitor thapsigargin leads to formation of anandamide, and subsequent TRPV1-dependent Ca2+ influx in transfected cells and sensory neurons of rat dorsal root ganglia (DRG). Anandamide metabolism and efflux from the cell tonically limit TRPV1-mediated Ca2+ entry. In DRG neurons, this mechanism was found to lead to TRPV1-mediated currents that were enhanced by selective blockade of anandamide cellular efflux. Thus, endogenous anandamide is formed on stimulation of metabotropic receptors coupled to the phospholipase C/inositol 1,4,5-triphosphate pathway and then signals to TRPV1 channels. This novel intracellular function of anandamide may precede its action at cannabinoid receptors, and might be relevant to its control over neurotransmitter release.
Collapse
Affiliation(s)
- Mario van der Stelt
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli (NA), Italy
| | - Marcello Trevisani
- Department of Experimental Medicine and Clinical Medicine, Pharmacology Unit, University of Ferrara, Ferrara, Italy
| | - Vittorio Vellani
- Dipartimento di Scienze Biomediche, Universita' di Modena e Reggio Emilia, Modena, Italy
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Luciano De Petrocellis
- Istitute of Cibernetica Eduardo Caianiello, Consiglio Nazionale delle Ricerche, Pozzuoli (NA), Italy
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli (NA), Italy
- Istitute of Cibernetica Eduardo Caianiello, Consiglio Nazionale delle Ricerche, Pozzuoli (NA), Italy
| | - Barbara Campi
- Department of Experimental Medicine and Clinical Medicine, Pharmacology Unit, University of Ferrara, Ferrara, Italy
| | - Peter McNaughton
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Piero Geppetti
- Department of Experimental Medicine and Clinical Medicine, Pharmacology Unit, University of Ferrara, Ferrara, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli (NA), Italy
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy. Tel.: +39 081 8675093; Fax: +39 081 8041770; E-mail:
| |
Collapse
|
32
|
Evans JF, Shen CL, Pollack S, Aloia JF, Yeh JK. Adrenocorticotropin evokes transient elevations in intracellular free calcium ([Ca2+]i) and increases basal [Ca2+]i in resting chondrocytes through a phospholipase C-dependent mechanism. Endocrinology 2005; 146:3123-32. [PMID: 15802497 DOI: 10.1210/en.2004-1612] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Both clinical and in vitro evidence points to the involvement of the melanocortin peptide, ACTH, in the terminal differentiation of chondrocytes. Terminal differentiation along the endochondral pathway is responsible for linear growth, but also plays a role in osteoarthritic cartilage degeneration. Chondrocyte terminal differentiation is associated with an incremental increase in chondrocyte basal intracellular free calcium ([Ca(2+)](i)), and ACTH agonism of melanocortin receptors is known to mobilize [Ca(2+)](i.) Using differentiated resting chondrocytes highly expressing type II collagen and aggrecan, we examined the influence of both ACTH and dexamethasone treatment on matrix gene transcription and [Ca(2+)](i). Resting chondrocytes treated concurrently with dexamethasone and ACTH expressed matrix gene transcripts in a pattern consistent with that of rapid terminal differentiation. Using the fluorescent Ca(2+) indicator, fura-2, we determined that ACTH evokes transient increases in [Ca(2+)](i) and elevates basal Ca(2+) levels in resting chondrocytes. The transient increases were initiated intracellularly, were abrogated by the phospholipase C-specific inhibitor, U73122, and were partly attenuated by myo-inositol 1,4,5-triphosphate receptor inhibition via 10 mm caffeine. The initial intracellular release also resulted in store-operated calcium entry, presumably through store-operated channels. Dexamethasone priming increased both the initial ACTH-evoked [Ca(2+)](i) release and the subsequent store-operated calcium entry. These data demonstrate roles for ACTH and glucocorticoid in the regulation of chondrocyte terminal differentiation. Because the actions of ACTH are mediated through known G protein-coupled receptors, the melanocortin receptors, these data may provide a new therapeutic target in the treatment of growth deficiencies and cartilage degeneration.
Collapse
Affiliation(s)
- Jodi F Evans
- Department of Medicine, Winthrop University Hospital, Mineola, New York 11501, USA.
| | | | | | | | | |
Collapse
|
33
|
Gill DL, Patterson RL. Toward a consensus on the operation of receptor-induced calcium entry signals. Sci Signal 2004; 2004:pe39. [PMID: 15280581 DOI: 10.1126/stke.2432004pe39] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Receptor-induced Ca2+ signals involve both Ca2+ release from intracellular stores and extracellular Ca2+ entry across the plasma membrane. The channels mediating Ca2+ entry and the mechanisms controlling their function remain largely a mystery. Here we critically assess current views on the Ca2+ entry process and consider certain modifications to the widely held hypothesis that Ca2+ store emptying is the fundamental trigger for receptor-induced Ca2+ entry channels. Under physiological conditions, receptor-induced store depletion may be quite limited. A number of distinct channel activities appear to mediate receptor-induced Ca2+ entry, and their activation is observed to occur through quite diverse coupling processes.
Collapse
Affiliation(s)
- Donald L Gill
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | |
Collapse
|