1
|
Cappelletti P, Filareti M, Masuelli L, Bei R, Hassanzadeh K, Corbo M, Feligioni M. Syntaxin-1a and SNAP-25 expression level is increased in the blood samples of ischemic stroke patients. Sci Rep 2022; 12:14483. [PMID: 36008522 PMCID: PMC9411545 DOI: 10.1038/s41598-022-18719-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022] Open
Abstract
The interest for the discovery of blood biomarkers for several neurological disorders, including Ischemic Stroke (IS), is growing and their identification in blood samples would be revolutionary allowing a fast and better pathology prediction or outcome and to collect information on patient recovery. The increased permeability of the blood-brain barrier, following a brain infarct, allows the detection of brain proteins in the blood flow. In this work, we analyzed the expression levels of two synaptic proteins Syntaxin (STX)-1a and Synaptosomal Associated Protein, 25 kDa (SNAP-25), in Peripheral Blood Mononuclear Cell (PBMC), serum and in Neuronal Derived Extracellular vesicles (NDEs) of IS patients, age and sex matched healthy control (HC) and younger HC (Y-HC). Interestingly, we identified STX-1a protein in the cytoplasm of PBMC and both STX-1a and SNAP-25 expression levels were significantly augmented in all IS patient's blood fractions compared to control subjects. In addition, STX-1a blood levels correlated with the IS clinical scales National Institutes of Health Stroke Scale (NIH-SS) and the modified Barthel Index (BI). These results prompted us to speculate that STX-1a and SNAP-25 hematic fluctuations depict the brain damage after an ischemic attack and that their hematic detection could represent a novel and accessible IS biomarkers.
Collapse
Affiliation(s)
- Pamela Cappelletti
- Department of Neuro-Rehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Melania Filareti
- Department of Neuro-Rehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Kambiz Hassanzadeh
- European Brain Research Institute (EBRI) Rita Levi Montalcini Foundation, Viale Regina Elena 295, 00161, Rome, Italy.,Fondazione Pisana per la Scienza (FPS), Pisa, Italy
| | - Massimo Corbo
- Department of Neuro-Rehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Marco Feligioni
- Department of Neuro-Rehabilitation Sciences, Casa Cura Policlinico, Milan, Italy. .,European Brain Research Institute (EBRI) Rita Levi Montalcini Foundation, Viale Regina Elena 295, 00161, Rome, Italy.
| |
Collapse
|
2
|
Wang LF, Ling DY, Huang MX, Tao LW, Tong QX, Hou Y, Li H, Chen Z, Zhang BZ, Lu HT, Wang YF, Zhang XG. Influence of atherosclerosis on the molecular expression of the TRPC1/BK signal complex in the aortic smooth muscles of mice. Exp Ther Med 2022; 23:4. [PMID: 34815756 PMCID: PMC8593874 DOI: 10.3892/etm.2021.10926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/08/2021] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis (AS) is one a disease that seriously endangers human health. Previous studies have demonstrated that transient receptor potential channel-1 (TRPC1)/large conductance Ca2+ activated K+ channel (BK) signal complex is widely distributed in arteries. Therefore, it was hypothesized that TRPC1-BK signal complex may be a new target for the treatment of AS-related diseases. Apolipoprotein E-/- (ApoE-/-) mice were used to establish an atherosclerotic animal model in the present study, and the association between AS and the TRPC1-BK signal complex was examined. The present study aimed to compare the differences in the expression levels of mRNAs and proteins of the TRPC1-BK signal complex expressed in the aortic vascular smooth muscle tissue, between mice with AS and control mice. There were 10 mice in each group. Reverse transcription PCR, western blotting and immunohistochemistry were used to detect the differences in the mRNA and protein expression levels of TRPC1, BKα (the α subunit of BK) and BKβ1 (the β1 subunit of BK). The mRNA expression level of TRPC1 in AS model mice was significantly higher compared with that in the control group (P<0.05). However, the mRNA expression levels of BKα and BKβ1 were lower compared with those in the controls (both P<0.01). The mice in the ApoE-/- group successfully developed AS. In this group, the protein expression level of TRPC1 was significantly higher than that in the control group (P<0.01), while the protein expression levels of BKα and BKβ1 were lower compared with those in the control group (P<0.01 and P<0.05, respectively). Collectively, it was identified that the protein and mRNA expression levels of the TRPC1/BK signal complex in the aortic vascular smooth muscle tissue could be influenced by the development of AS in mice. Hence, the TRPC1/BK signal complex may be a potential therapeutic target for the prevention and treatment of AS-related complications in the future.
Collapse
Affiliation(s)
- Lian-Fa Wang
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Dong-Yun Ling
- Department of Cardiology, The Second People's Hospital of Hefei City, Hefei, Anhui 230011, P.R. China
| | - Meng-Xun Huang
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Li-Wei Tao
- Department of Cardiothoracic Surgery, The Second People's Hospital of Fuyang City, Fuyang, Anhui 236000, P.R. China
| | - Quan-Xiu Tong
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Yong Hou
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Hua Li
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Zhen Chen
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Bang-Zhu Zhang
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Hong-Tao Lu
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Yun-Fei Wang
- Department of Cardiology, The 901st Hospital of Joint Logistics Support Force of PLA, Hefei, Anhui 230031, P.R. China
| | - Xian-Ge Zhang
- Institute of Public Health and Nursing Research, Department of Healthcare Management, University of Bremen, 28359 Bremen, Germany
| |
Collapse
|
3
|
Sharma A, Ramena GT, Elble RC. Advances in Intracellular Calcium Signaling Reveal Untapped Targets for Cancer Therapy. Biomedicines 2021; 9:1077. [PMID: 34572262 PMCID: PMC8466575 DOI: 10.3390/biomedicines9091077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Intracellular Ca2+ distribution is a tightly regulated process. Numerous Ca2+ chelating, storage, and transport mechanisms are required to maintain normal cellular physiology. Ca2+-binding proteins, mainly calmodulin and calbindins, sequester free intracellular Ca2+ ions and apportion or transport them to signaling hubs needing the cations. Ca2+ channels, ATP-driven pumps, and exchangers assist the binding proteins in transferring the ions to and from appropriate cellular compartments. Some, such as the endoplasmic reticulum, mitochondria, and lysosomes, act as Ca2+ repositories. Cellular Ca2+ homeostasis is inefficient without the active contribution of these organelles. Moreover, certain key cellular processes also rely on inter-organellar Ca2+ signaling. This review attempts to encapsulate the structure, function, and regulation of major intracellular Ca2+ buffers, sensors, channels, and signaling molecules before highlighting how cancer cells manipulate them to survive and thrive. The spotlight is then shifted to the slow pace of translating such research findings into anticancer therapeutics. We use the PubMed database to highlight current clinical studies that target intracellular Ca2+ signaling. Drug repurposing and improving the delivery of small molecule therapeutics are further discussed as promising strategies for speeding therapeutic development in this area.
Collapse
Affiliation(s)
- Aarushi Sharma
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Grace T. Ramena
- Department of Aquaculture, University of Arkansas, Pine Bluff, AR 71601, USA;
| | - Randolph C. Elble
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| |
Collapse
|
4
|
STIM-TRP Pathways and Microdomain Organization: Contribution of TRPC1 in Store-Operated Ca 2+ Entry: Impact on Ca 2+ Signaling and Cell Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:159-188. [PMID: 28900914 DOI: 10.1007/978-3-319-57732-6_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Store-operated calcium entry (SOCE) is a ubiquitous Ca2+ entry pathway that is activated in response to depletion of ER-Ca2+ stores and critically controls the regulation of physiological functions in a wide variety of cell types. The transient receptor potential canonical (TRPC) channels (TRPCs 1-7), which are activated by stimuli leading to PIP2 hydrolysis, were first identified as molecular components of SOCE channels. While TRPC1 was associated with SOCE and regulation of function in several cell types, none of the TRPC members displayed I CRAC, the store-operated current identified in lymphocytes and mast cells. Intensive search finally led to the identification of Orai1 and STIM1 as the primary components of the CRAC channel. Orai1 was established as the pore-forming channel protein and STIM1 as the ER-Ca2+ sensor protein involved in activation of Orai1. STIM1 also activates TRPC1 via a distinct domain in its C-terminus. However, TRPC1 function depends on Orai1-mediated Ca2+ entry, which triggers recruitment of TRPC1 into the plasma membrane where it is activated by STIM1. TRPC1 and Orai1 form distinct store-operated Ca2+ channels that regulate specific cellular functions. It is now clearly established that regulation of TRPC1 trafficking can change plasma membrane levels of the channel, the phenotype of the store-operated Ca2+ current, as well as pattern of SOCE-mediated [Ca2+]i signals. Thus, TRPC1 is activated downstream of Orai1 and modifies the initial [Ca2+]i signal generated by Orai1. This review will highlight current concepts of the activation and regulation of TRPC1 channels and its impact on cell function.
Collapse
|
5
|
Ong HL, de Souza LB, Ambudkar IS. Role of TRPC Channels in Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:87-109. [DOI: 10.1007/978-3-319-26974-0_5] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
6
|
Berna-Erro A, Jardín I, Smani T, Rosado JA. Regulation of Platelet Function by Orai, STIM and TRP. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:157-81. [PMID: 27161229 DOI: 10.1007/978-3-319-26974-0_8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Agonist-induced changes in cytosolic Ca(2+) concentration ([Ca(2+)]c) are central events in platelet physiology. A major mechanism supporting agonist-induced Ca(2+) signals is store-operated Ca(2+) entry (SOCE), where the Ca(2+) sensor STIM1 and the channels of the Orai family, as well as TRPC members are the key elements. STIM1-dependent SOCE plays a major role in collagen-stimulated Ca(2+) signaling, phosphatidylserine exposure and thrombin generation. Furthermore, studies involving Orai1 gain-of-function mutants and platelets from Orai1-deficient mice have revealed the importance of this channel in thrombosis and hemostasis to those found in STIM1-deficient mice indicating that SOCE might play a prominent role in thrombus formation. Moreover, increase in TRPC6 expression might lead to thrombosis in humans. The role of STIM1, Orai1 and TRPCs, and thus SOCE, in thrombus formation, suggests that therapies directed against SOCE and targeting these molecules during cardiovascular and cerebrovascular events could significantly improve traditional anti-thrombotic treatments.
Collapse
Affiliation(s)
- Alejandro Berna-Erro
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Isaac Jardín
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Cáceres, 10003, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysic, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, 41013, Spain
| | - Juan A Rosado
- Departamento de Fisiología, University of Extremadura, Cáceres, Spain.
| |
Collapse
|
7
|
Ong HL, Ambudkar IS. Molecular determinants of TRPC1 regulation within ER–PM junctions. Cell Calcium 2015; 58:376-86. [DOI: 10.1016/j.ceca.2015.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 11/30/2022]
|
8
|
Chen F, Li Q, Gu M, Li X, Yu J, Zhang YB. Identification of a Mutation in FGF23 Involved in Mandibular Prognathism. Sci Rep 2015; 5:11250. [PMID: 26059428 PMCID: PMC4462018 DOI: 10.1038/srep11250] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 05/18/2015] [Indexed: 12/30/2022] Open
Abstract
Mandibular prognathism (MP) is a severe maxillofacial disorder with undetermined genetic background. We collected a Chinese pedigree with MP which involved in 23 living members of 4 generations. Genome-wide linkage analysis were carried out to obtain the information in this family and a new MP-susceptibility locus, 12pter-p12.3 was identified. Whole-exome sequencing identified a novel heterozygous mutation in fibroblast growth factor (FGF) 23 (; p.A12D) which well segregated with MP in this pedigree within the locus. The mutation was also detected in 3 cases out of 65 sporadic MP patients, but not in any of the 342 control subjects. The p.A12D mutation may disrupt signal peptide function and inhibit secretory in FGF23. Furthermore, mutant FGF23 was overexpressed in 293T cells, increased cytoplasmic accumulation was observed compared with the wild type. We have discovered that c.35C>A mutation in FGF23 strongly associated with MP, which expand our understanding of the genetic contribution to MP pathogenesis.
Collapse
Affiliation(s)
- Fengshan Chen
- Laboratory of Oral Biomedical Science and Translational Medicine, School and Hospital of Stomatology, Tongji University, Shanghai, P. R. China
| | - Qin Li
- Laboratory of Oral Biomedical Science and Translational Medicine, School and Hospital of Stomatology, Tongji University, Shanghai, P. R. China
| | - Mingliang Gu
- Beijing Institute of Genomics, Chinese Academy of Sciences and Key Laboratory of GenomeScience and Information, Chinese Academy of Sciences, Beijing, P. R. China
| | - Xin Li
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, P. R. China
| | - Jun Yu
- Beijing Institute of Genomics, Chinese Academy of Sciences and Key Laboratory of GenomeScience and Information, Chinese Academy of Sciences, Beijing, P. R. China
| | - Yong-Biao Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences and Key Laboratory of GenomeScience and Information, Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
9
|
Redondo PC, Rosado JA. Store-operated calcium entry: unveiling the calcium handling signalplex. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 316:183-226. [PMID: 25805125 DOI: 10.1016/bs.ircmb.2015.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Store-operated Ca(2+) entry (SOCE) is an important mechanism for Ca(2+) influx in non-excitable cells, also present in excitable cells. The activation of store-operated channels (SOCs) is finely regulated by the filling state of the intracellular agonist-sensitive Ca(2+) compartments, and both, the mechanism of sensing the Ca(2+) stores and the nature and functional properties of the SOCs, have been a matter of intense investigation and debate. The identification of STIM1 as the endoplasmic reticulum Ca(2+) sensor and both Orai1, as the pore-forming subunit of the channels mediating the Ca(2+)-selective store-operated current, and the members of the TRPC subfamily of proteins, as the channels mediating the cation-permeable SOCs, has shed new light on the underlying events. This review summarizes the initial hypothesis and the current advances on the mechanism of activation of SOCE.
Collapse
Affiliation(s)
- Pedro C Redondo
- Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Cáceres, Spain
| |
Collapse
|
10
|
de Souza LB, Ambudkar IS. Trafficking mechanisms and regulation of TRPC channels. Cell Calcium 2014; 56:43-50. [PMID: 25012489 DOI: 10.1016/j.ceca.2014.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 02/06/2023]
Abstract
TRPC channels are Ca(2+)-permeable cation channels which are regulated downstream from receptor-coupled PIP2 hydrolysis. These channels contribute to a wide variety of cellular functions. Loss or gain of channel function has been associated with dysfunction and aberrant physiology. TRPC channel functions are influenced by their physical and functional interactions with numerous proteins that determine their regulation, scaffolding, trafficking, as well as their effects on the downstream cellular processes. Such interactions also compartmentalize the Ca(2+) signals arising from TRPC channels. A large number of studies demonstrate that trafficking is a critical mode by which plasma membrane localization and surface expression of TRPC channels are regulated. This review will provide an overview of intracellular trafficking pathways as well as discuss the current state of knowledge regarding the mechanisms and components involved in trafficking of the seven members of the TRPC family (TRPC1-TRPC7).
Collapse
Affiliation(s)
- Lorena Brito de Souza
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States.
| | - Indu S Ambudkar
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
11
|
Ong HL, de Souza LB, Cheng KT, Ambudkar IS. Physiological functions and regulation of TRPC channels. Handb Exp Pharmacol 2014; 223:1005-34. [PMID: 24961978 DOI: 10.1007/978-3-319-05161-1_12] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The TRP-canonical (TRPC) subfamily, which consists of seven members (TRPC1-TRPC7), are Ca(2+)-permeable cation channels that are activated in response to receptor-mediated PIP2 hydrolysis via store-dependent and store-independent mechanisms. These channels are involved in a variety of physiological functions in different cell types and tissues. Of these, TRPC6 has been linked to a channelopathy resulting in human disease. Two key players of the store-dependent regulatory pathway, STIM1 and Orai1, interact with some TRPC channels to gate and regulate channel activity. The Ca(2+) influx mediated by TRPC channels generates distinct intracellular Ca(2+) signals that regulate downstream signaling events and consequent cell functions. This requires localization of TRPC channels in specific plasma membrane microdomains and precise regulation of channel function which is coordinated by various scaffolding, trafficking, and regulatory proteins.
Collapse
Affiliation(s)
- Hwei Ling Ong
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | |
Collapse
|
12
|
G protein-coupled receptor signalling potentiates the osmo-mechanical activation of TRPC5 channels. Pflugers Arch 2013; 466:1635-46. [DOI: 10.1007/s00424-013-1392-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/04/2013] [Accepted: 10/19/2013] [Indexed: 10/26/2022]
|
13
|
Ryu HJ, Kim JE, Kim YJ, Kim JY, Kim WIL, Choi SY, Kim MJ, Kang TC. Endothelial transient receptor potential conical channel (TRPC)-3 activation induces vasogenic edema formation in the rat piriform cortex following status epilepticus. Cell Mol Neurobiol 2013; 33:575-85. [PMID: 23529532 DOI: 10.1007/s10571-013-9931-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 03/18/2013] [Indexed: 11/29/2022]
Abstract
Transient receptor potential canonical channel (TRPC) is a nonselective cation channel permeable to Ca(2+), which express in many cell types, including neurons. However the alterations in TRPC receptor expressions in response to status epilepticus (SE) have not been explored. Therefore, the present study was designated to elucidate the roles of TRPC3 in neuronal death and vasogenic edema within the rat piriform cortex (PC) following SE. In non-SE animals, TRPC3 immunoreactivity was abundantly detected in the PC. Following SE, TRPC3 immunoreactivity was increased in neurons. Furthermore, TRPC3 expression was detected in endothelial cells that did not contain it in non-SE animals. Loss of SMI-71 (a blood-brain barrier antigen) immunoreactivity was also observed in TRPC3 positive endothelial cells. In addition, FJB positive neurons and vasogenic edema were noticeably detected in the PC. To directly determine whether TRPC3 activation is correlated to SE-induced vasogenic edema formation and neuronal damages in the PC, the effect of Pyr-3 (a TRPC3 antagonist) on SE-induced insults were investigated. Pyr-3 infusion effectively attenuated vasogenic edema in the PC as compared to the vehicle. Therefore, our findings indicate that TRPC3 activation/overexpression induced by SE may involve BBB disruption and neuronal damages in the rat PC following SE. Therefore, the present study was TRPC3 may play an important role in SE-induced vasogenic edema formation through BBB disruptions in the rat PC.
Collapse
Affiliation(s)
- Hea Jin Ryu
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Kim DS, Ryu HJ, Kim JE, Kang TC. The reverse roles of transient receptor potential canonical channel-3 and -6 in neuronal death following pilocarpine-induced status epilepticus. Cell Mol Neurobiol 2013; 33:99-109. [PMID: 22926417 DOI: 10.1007/s10571-012-9875-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 08/14/2012] [Indexed: 12/13/2022]
Abstract
Transient receptor potential canonical channel (TRPC) is a nonselective cation channel permeable to Ca(2+), which is expressed in many cell types, including neurons. However, the alterations in TRPC receptor expressions in response to status epilepticus (SE) have not been explored. Therefore, the present study was designated to elucidate the roles of TRPC3 and TRPC6 in neuronal death following SE. In non-SE animals, TRPC3 and TRPC6 immunoreactivity was abundantly detected in the dendrites of pyramidal cells and the cell bodies of dentate granule cells. Following SE, TRPC3 expression was significantly elevated in CA1-, CA3 pyramidal cells, and dentate granule cells, while TRPC6 expression was reduced in these regions. Pyrazole-3 (a TRPC3 inhibitor) effectively prevented up-regulation of neuronal TRPC3 expression induced by SE. Hyperforin (a TRPC6 activator) effectively prevented down-regulation of neuronal TRPC6 expression induced by SE. In addition, both Pyr3 and hyperforin effectively protected neuronal damages from SE. Therefore, the present study yields novel information regarding the role of TRPC3 and 6 in epileptogenic insults and suggests that TRPC 3 and 6 may be involved in neurodegeneration following SE.
Collapse
Affiliation(s)
- Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam-Do, 330-090, South Korea
| | | | | | | |
Collapse
|
15
|
Thrombin-stimulated discharge of calcium stores in human platelets: analysis of experimental data. Arch Biochem Biophys 2012; 526:78-83. [PMID: 22846427 DOI: 10.1016/j.abb.2012.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 11/21/2022]
Abstract
The purpose of this research was to analyze experimental data concerning thrombin-stimulated discharge of calcium stores in human platelets contained in calcium-free medium in view of better understanding the mechanisms involved in calcium fluxes. The model curves are reasonably close to experimental data; the parameters of the models are related to the properties of the entities responsible for control or maintenance of cytosolic calcium concentration. It has been shown that: (a) time-course of calcium concentration in cytosol of human platelets can be acceptably modeled on the basis of reasonable assumptions concerning agonist stimulated calcium redistribution in cellular compartments; (b) those assumptions are of fundamental importance for the model (c) some parameters of the model (taken arbitrarily) cannot be estimated independently of others from fitting the model to experimental data available; (d) special experiments are necessary to determine the unknown parameters; (e) agonist-stimulated change of the permeability of endomembrane of calcium stores can be regarded as a pulse of the permeability; it can be modeled as a sequence of transitions of the system from inactive to active and to inactive state again.
Collapse
|
16
|
Furuya TK, Silva PNO, Payão SLM, Bertolucci PHF, Rasmussen LT, De Labio RW, Braga ILS, Chen ES, Turecki G, Mechawar N, Mill J, Smith MAC. Analysis of SNAP25 mRNA expression and promoter DNA methylation in brain areas of Alzheimer's Disease patients. Neuroscience 2012; 220:41-6. [PMID: 22732502 DOI: 10.1016/j.neuroscience.2012.06.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 06/13/2012] [Accepted: 06/14/2012] [Indexed: 10/28/2022]
Abstract
Alzheimer's Disease (AD) is the most common cause of dementia in elderly people. The presynaptic terminal is an important site of pathological changes in AD, leading to synaptic loss in specific brain regions, such as in the cortex and hippocampus. In this study, we investigated synaptosomal-associated protein, 25-kDa (SNAP25) mRNA levels and promoter DNA methylation in post mortem brain tissues (entorhinal and auditory cortices and hippocampus) from healthy elderly and AD subjects as well as in peripheral blood leukocytes of young, healthy elderly and AD patients. mRNA quantification was performed by quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR) using the ΔΔC(T) method and promoter DNA methylation was quantified by mass spectrometry using the Sequenom EpiTYPER platform. We observed a significant decrease in SNAP25 expression in AD across all the three brain regions in relation to the healthy elderly subjects, suggesting impairment in synaptic function. The changes in the auditory cortex reflected those observed in the hippocampus and entorhinal cortex, the primary areas affected in AD. However, no AD-associated differences in SNAP25 promoter DNA methylation were observed suggesting that other mechanisms may be involved in mediating the observed gene expression changes.
Collapse
Affiliation(s)
- T K Furuya
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo (UNIFESP), São Paulo-SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Juška A. Calcium fluxes into and out of cytosol in human platelets: analysis of experimental data. Biochem Biophys Res Commun 2011; 412:537-42. [PMID: 21798249 DOI: 10.1016/j.bbrc.2011.07.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 07/11/2011] [Indexed: 12/11/2022]
Abstract
The purpose of this research was to analyse experimental data concerning cytosolic calcium concentration in view of the mechanisms involved in calcium fluxes in human platelets. The parameters of model curves are related to the properties of the entities responsible for control or maintenance of cytosolic calcium concentration. It has been shown that: (a) biphasicity of increase in cytosolic calcium concentration caused by inhibition of SERCAs either by TBHQ and TG or by TG alone is related to fast and slow discharge of acidic calcium stores and DTS; (b) biphasicity of decline in cytosolic calcium concentration after its rise caused by stimulation of platelets by the agonists is related to non-synchronous extrusion of calcium by PMCA and NCX; (c) NCX is active only in calcium containing medium: calcium ion(s) are necessary to be bound to the site(s) located on the medium-facing side of the (macro)molecule; (d) PMCA is likely to be activated either by binding calcium ion(s) to the site(s) located on its cytosol-facing side or by unbinding identical ion(s) from the site(s) on its medium-facing side.
Collapse
Affiliation(s)
- Alfonsas Juška
- Vilniaus Gedimino technikos universitetas, Vilnius, Lithuania.
| |
Collapse
|
18
|
Popoff MR, Poulain B. Bacterial toxins and the nervous system: neurotoxins and multipotential toxins interacting with neuronal cells. Toxins (Basel) 2010; 2:683-737. [PMID: 22069606 PMCID: PMC3153206 DOI: 10.3390/toxins2040683] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 03/18/2010] [Accepted: 04/07/2010] [Indexed: 12/13/2022] Open
Abstract
Toxins are potent molecules used by various bacteria to interact with a host organism. Some of them specifically act on neuronal cells (clostridial neurotoxins) leading to characteristics neurological affections. But many other toxins are multifunctional and recognize a wider range of cell types including neuronal cells. Various enterotoxins interact with the enteric nervous system, for example by stimulating afferent neurons or inducing neurotransmitter release from enterochromaffin cells which result either in vomiting, in amplification of the diarrhea, or in intestinal inflammation process. Other toxins can pass the blood brain barrier and directly act on specific neurons.
Collapse
Affiliation(s)
- Michel R. Popoff
- Neurotransmission et Sécrétion Neuroendocrine, CNRS UPR 2356 IFR 37 - Neurosciences, Centre de Neurochimie, 5, rue Blaise Pascal, F-67084 STRASBOURG cedex, France;
- Author to whom correspondence should be addressed;
| | | |
Collapse
|
19
|
Tam HW, Huang YC, Tam MF. A cost-effective device for the rapid transfer of gel-separated proteins onto membranes. Anal Biochem 2009; 386:123-5. [PMID: 19094958 DOI: 10.1016/j.ab.2008.11.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 11/14/2008] [Accepted: 11/14/2008] [Indexed: 10/21/2022]
Abstract
We describe here the fabrication of a cost-effective semi-dry blotting apparatus for the transfer of proteins onto membranes. Graphite sheets were used as electrodes. Protein mixtures were separated on NuPAGE 4% to 12% polyacrylamide gradient gels. With a Tris-bicine buffer, we demonstrated that close to 80% of the proteins with apparent molecular mass of 80kDa or less were removed from the gels after 8min of blotting. The process is much faster than the techniques reported previously in the literature.
Collapse
Affiliation(s)
- Hann W Tam
- Institute of Molecular Biology, Academia Sinica, NanKang Taipei, Taiwan, ROC
| | | | | |
Collapse
|
20
|
Woodard GE, Salido GM, Rosado JA. Enhanced exocytotic-like insertion of Orai1 into the plasma membrane upon intracellular Ca2+ store depletion. Am J Physiol Cell Physiol 2008; 294:C1323-31. [PMID: 18400989 DOI: 10.1152/ajpcell.00071.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Ca+ release-activated Ca2+ (CRAC) channels are activated when free Ca2+ concentration in the intracellular stores is substantially reduced and mediate sustained Ca2+ entry. Recent studies have identified Orai1 as a CRAC channel subunit. Here we demonstrate that passive Ca2+ store depletion using the inhibitor of the sarcoendoplasmic reticulum Ca2+-ATPase, thapsigargin (TG), enhances the surface expression of Orai1, a process that depends on rises in cytosolic free Ca2+ concentration, as demonstrated in cells loaded with dimethyl BAPTA, an intracellular Ca2+ chelator that prevented TG-evoked cytosolic free Ca2+ concentration elevation. Similar results were observed with a low concentration of carbachol. Cleavage of the soluble N-ethylmaleimide-sensitive-factor attachment protein receptor, synaptosomal-assiciated protein-25 (SNAP-25), with botulinum neurotoxin A impaired TG-induced increase in the surface expression of Orai1. In addition, SNAP-25 cleaving by botulinum neurotoxin A reduces the maintenance but not the initial stages of store-operated Ca2+ entry. In aggregate, these findings demonstrate that store depletion enhances Orai1 plasma membrane expression in an exocytotic manner that involves SNAP-25, a process that contributes to store-dependent Ca2+ entry.
Collapse
Affiliation(s)
- Geoffrey E Woodard
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
21
|
Redondo PC, Jardin I, Lopez JJ, Salido GM, Rosado JA. Intracellular Ca2+ store depletion induces the formation of macromolecular complexes involving hTRPC1, hTRPC6, the type II IP3 receptor and SERCA3 in human platelets. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1783:1163-76. [PMID: 18191041 DOI: 10.1016/j.bbamcr.2007.12.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 12/11/2007] [Accepted: 12/11/2007] [Indexed: 11/26/2022]
Abstract
Endogenously expressed human canonical transient receptor potential 1 (hTRPC1) and human canonical transient receptor potential 6 (hTRPC6) have been shown to play a role in store-operated Ca2+ entry (SOCE) in human platelets, where two mechanisms for SOCE, regulated by the dense tubular system (DTS) or the acidic granules, have been identified. In cells preincubated for 1 min with 100 microM flufenamic acid we show that hTRPC6 is involved in SOCE activated by both mechanisms, as demonstrated by selective depletion of the DTS or the acidic stores, using thapsigargin (TG) (10 nM) or 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ) (20 microM), respectively, although it is more relevant after acidic store depletion. Co-immunoprecipitation experiments indicated that depletion of both stores separately results in time-dependent interaction between hTRPC1 and hTRPC6, and also between both hTRPCs and the type II IP3 receptor (IP3RII). The latter was greater after treatment with TG. TBHQ-induced coupling between hTRPC1 and 6 was transient and decreased after 30s of treatment, while that induced by TG increased for at least 3 min. TBHQ induced association between SERCA3, located in the acidic stores, hTRPC1, hTRPC6 and Orai1. TBHQ also evoked coupling between SERCA3 and IP3RII, presumably located in the DTS, thus suggesting interplay between both Ca2+ stores. Similarly, TG induces the interaction of SERCA2b with hTRPC1 and 6 and the IP3RII. The interactions between hTRPC1, hTRPC6, IP3RII and SERCA3 were impaired by disruption of the microtubules, supporting a role for microtubules in Ca2+ homeostasis. In conclusion, the present data demonstrate for the first time that hTRPC1, hTRPC6, IP3RII and SERCA3 are parts of a macromolecular protein complex activated by depletion of the intracellular Ca2+ stores in human platelets.
Collapse
Affiliation(s)
- Pedro C Redondo
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3EG Cambridge, UK.
| | | | | | | | | |
Collapse
|
22
|
Jardín I, López JJ, Salido GM, Rosado JA. Functional relevance of the de novo coupling between hTRPC1 and type II IP3 receptor in store-operated Ca2+ entry in human platelets. Cell Signal 2007; 20:737-47. [PMID: 18249094 DOI: 10.1016/j.cellsig.2007.12.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 12/13/2007] [Accepted: 12/13/2007] [Indexed: 12/16/2022]
Abstract
Store-operated Ca2+ entry (SOCE), a major mechanism for Ca2+ entry in non-excitable cells, is regulated by the filling state of the intracellular Ca2+ stores. We have previously reported that a de novo conformational coupling between the type II IP3 receptor (IP3RII) and hTRPC1 channel occurs after depletion of the intracellular Ca2+ stores in human platelets, which might be involved in the activation of SOCE in these cells. Here we present for the first time direct evidence for the functional relevance of the coupling between hTRPC1 and IP3RII in SOCE in human platelets. Our data suggest that at least two pathways may contribute to SOCE in these cells. An early component, insensitive to cytochalasin D (Cyt D), is followed by a late component which is sensitive to Cyt D. Introduction of a peptide corresponding to IP3RII(317-334) (IP3BD-peptide(317-334)) in the cells by electrotransjection impairs the coupling between hTRPC1 and IP3RII but not the interaction between hTRPC1 and STIM1 induced by store depletion. Coimmunoprecipitation experiments indicated that endogenously expressed hTRPC1 interacts with the IP3BD-peptide(317-334). Electrotransjection of cells with IP3BD-peptide(317-334), significantly attenuated the late stage of Ca2+ and Mn2+ entry induced by 10 nM thapsigargin (TG) or 20 microM 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ), providing evidence for a functional role of the de novo coupling between hTRPC1 and IP3RII in the activation of SOCE in human platelets.
Collapse
Affiliation(s)
- Isaac Jardín
- Department of Physiology (Cellular Physiology Research Group), University of Extremadura, 10071 Cáceres, Spain
| | | | | | | |
Collapse
|
23
|
Jardin I, Ben Amor N, Hernández-Cruz JM, Salido GM, Rosado JA. Involvement of SNARE proteins in thrombin-induced platelet aggregation: Evidence for the relevance of Ca2+ entry. Arch Biochem Biophys 2007; 465:16-25. [PMID: 17543880 DOI: 10.1016/j.abb.2007.04.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 04/27/2007] [Accepted: 04/29/2007] [Indexed: 10/23/2022]
Abstract
Thrombin induces platelet activation through a variety of intracellular mechanisms, including Ca(2+) mobilization. The protein of the exocytotic machinery SNAP-25, but not VAMPs, is required for store-operated Ca(2+) entry, the main mechanism for Ca(2+) influx in platelets. Hence, we have investigated the role of the SNAP-25 and VAMPs in thrombin-induced platelet aggregation. Platelet stimulation with thrombin or selective activation of thrombin receptors PAR-1, PAR-4 or GPIb-IX-V results in platelet aggregation that, except for GPIb-IX-V receptor, requires Ca(2+) entry for full activation. Depletion of the intracellular Ca(2+) stores using pharmacological tools was unable to induce aggregation except when cytosolic Ca(2+) concentration reached a critical level (around 1.5 microM). Electrotransjection of cells with anti-SNAP-25 antibody reduced thrombin-evoked platelet aggregation, while electrotransjection of anti-VAMP-1, -2 and -3 antibody had no effect. These findings support a role for SNAP-25 but not VAMP-1, -2 and -3 in platelet aggregation, which is likely mediated by the regulation of Ca(2+) mobilization in human platelets.
Collapse
Affiliation(s)
- Isaac Jardin
- Department of Physiology, Cellular Physiology Research Group, University of Extremadura, Av. Universidad s/n, Cáceres 10071, Spain
| | | | | | | | | |
Collapse
|
24
|
Suzuki F, Morishima S, Tanaka T, Muramatsu I. Snapin, a new regulator of receptor signaling, augments alpha1A-adrenoceptor-operated calcium influx through TRPC6. J Biol Chem 2007; 282:29563-73. [PMID: 17684020 DOI: 10.1074/jbc.m702063200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Activation of G(q)-protein-coupled receptors, including the alpha(1A)-adrenoceptor (alpha(1A)-AR), causes a sustained Ca(2+) influx via receptor-operated Ca(2+) (ROC) channels, following the transient release of intracellular Ca(2+). Transient receptor potential canonical (TRPC) channel is one of the candidate proteins constituting the ROC channels, but the precise mechanism linking receptor activation to increased influx of Ca(2+) via TRPCs is not yet fully understood. We identified Snapin as a protein interacting with the C terminus of the alpha(1A)-AR. In receptor-expressing PC12 cells, co-transfection of Snapin augmented alpha(1A)-AR-stimulated sustained increases in intracellular Ca(2+) ([Ca(2+)](i)) via ROC channels. By altering the Snapin binding C-terminal domain of the alpha(1A)-AR or by reducing cellular Snapin with short interfering RNA, the sustained increase in [Ca(2+)](i) in Snapin-alpha(1A)-AR co-expressing PC12 cells was attenuated. Snapin co-immunoprecipitated with TRPC6 and alpha(1A)-AR, and these interactions were augmented upon alpha(1A)-AR activation, increasing the recruitment of TRPC6 to the cell surface. Our data suggest a new receptor-operated signaling mechanism where Snapin links the alpha(1A)-AR to TRPC6, augmenting Ca(2+) influx via ROC channels.
Collapse
Affiliation(s)
- Fumiko Suzuki
- Division of Pharmacology, Department of Biochemistry and Bioinformative Sciences, School of Medicine, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui 910-1193, Japan
| | | | | | | |
Collapse
|
25
|
Cayouette S, Boulay G. Intracellular trafficking of TRP channels. Cell Calcium 2007; 42:225-32. [PMID: 17368756 DOI: 10.1016/j.ceca.2007.01.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 01/30/2007] [Accepted: 01/31/2007] [Indexed: 11/18/2022]
Abstract
Thirteen years ago, it was suggested that exocytotic insertion of store-operated channels into the plasma membrane lead to increased Ca(2+) entry in non-excitable cells upon G protein-coupled or tyrosine kinase receptor stimulation. Since the discovery of the TRP channel superfamily and their involvement in receptor-induced Ca(2+) entry, many studies have shown that different members of the TRP superfamily translocate into the plasma membrane upon stimulation. While the exact molecular mechanism by which TRP channels insert into the plasma membrane is unknown, TRP-binding proteins have been shown to directly regulate this trafficking. This review summarizes recent advances related to the mechanism of TRP channel trafficking, focusing on the role of TRP-binding proteins.
Collapse
Affiliation(s)
- Sylvie Cayouette
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | |
Collapse
|
26
|
London FS, Marcinkiewicz M, Walsh PN. PAR-1-stimulated factor IXa binding to a small platelet subpopulation requires a pronounced and sustained increase of cytoplasmic calcium. Biochemistry 2006; 45:7289-98. [PMID: 16752917 PMCID: PMC2533735 DOI: 10.1021/bi060294m] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We previously reported that only a subpopulation of PAR-1-stimulated platelets binds coagulation factor IXa, since confirmed by other laboratories. Since calcium changes have been implicated in exposure of procoagulant aminophospholipids, we have now examined calcium fluxes in this subpopulation by measuring fluorescence changes in Fura Red/AM-loaded platelets following PAR-1 stimulation. While fluorescence changes in all platelets indicated calcium release from internal stores and influx of external calcium, a subpopulation of platelets displayed a pronounced increase in calcium transients by 15 s and positive factor IXa binding by 2 min, with calcium transients sustained for 45 min. Pretreatment of platelets with Xestospongin C to inhibit IP3-mediated dense tubule calcium release, and the presence of impermeable calcium channel blockers nifedipine, SKF96365, or LaCl3, inhibited PAR-1-induced development of a subpopulation with pronounced calcium transients, factor IXa binding, and platelet support of FXa generation, suggesting the importance of both release of calcium from internal stores and influx of extracellular calcium. When platelets were stimulated in EDTA for 5-20 min before addition of calcium, factor IXa binding sites developed on a smaller subpopulation but with unchanged rate, indicating sustained opening of calcium channels and continued availability of signaling elements required for binding site exposure. While pretreatment of platelets with 100 microM BAPTA/AM (Kd 160 nM) had minimal effects, 100 microM 5,5'-dimethylBAPTA/AM (Kd 40 nM) completely inhibited the appearance and function of the platelet subpopulation, indicating the importance of minor increases of cytoplasmic calcium. We conclude that PAR-1-stimulated development of factor IXa binding sites in a subpopulation of platelets is dependent upon release of calcium from internal stores leading to sustained and pronounced calcium transients.
Collapse
Affiliation(s)
- Fredda S London
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA.
| | | | | |
Collapse
|
27
|
Goto S, Tamura N, Ishida H, Ruggeri ZM. Dependence of platelet thrombus stability on sustained glycoprotein IIb/IIIa activation through adenosine 5'-diphosphate receptor stimulation and cyclic calcium signaling. J Am Coll Cardiol 2005; 47:155-62. [PMID: 16386680 DOI: 10.1016/j.jacc.2005.08.055] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2005] [Revised: 07/24/2005] [Accepted: 08/01/2005] [Indexed: 12/16/2022]
Abstract
OBJECTIVES We sought to evaluate the mechanisms that support the stability of platelet aggregates on a thrombogenic surface exposed to flowing blood. BACKGROUND Activation of the membrane glycoprotein (GP) IIb/IIIa--mediated in part through the P2Y1 and P2Y12 adenosine 5'-diphosphate (ADP) receptors--is necessary for platelet aggregation. Platelets in growing thrombi exhibit cyclic calcium signal, suggesting that sustained activation may be required for thrombus stability. METHODS Blood was perfused over type I collagen fibrils at the wall shear rate of 1,500 s(-1). Three-dimensional visualization of platelet thrombi was obtained in real time with confocal microscopy. The intracytoplasmic Ca2+ concentration ([Ca2+]i) was measured in fluo-3AM-loaded platelets. RESULTS The height of platelet thrombi in control blood was 13.5 +/- 3.3 microm after 6 min, and increased to 16.3 +/- 4.5 microm (n = 8) after an additional 6 min. In contrast, the height was reduced to 5.4 +/- 2.2 and 3.3 +/- 1.3 microm, respectively (p < 0.01, n = 8), when the blood used in the second 6-min perfusion contained a P2Y1 (MRS2179) or P2Y12 (AR-C69931MX) inhibitor. The [Ca2+]i of platelets within forming thrombi oscillated between 212 +/- 38 nmol/l and 924 +/- 458 nmol/l, with cycles lasting 4.2 +/- 2.8 s that were inhibited completely by AR-C69931MX and partially by MRS2179. Accordingly, thrombi became unstable upon perfusion of blood containing the Ca2+ channel blocker, lanthanum chloride. Flow cytometric studies demonstrated that AR-C69931MX, MRS2179, and lanthanum chloride reduced monoclonal antibody PAC-1 binding to platelets, indicating a decrease of membrane-expressed activated GP IIb/IIIa. CONCLUSIONS Continuous P2Y1 and P2Y12 stimulation resulting in cyclic [Ca2+]i oscillations is required for maintaining the activation of GP IIb/IIIa needed for thrombus stability in flowing blood.
Collapse
Affiliation(s)
- Shinya Goto
- Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan.
| | | | | | | |
Collapse
|
28
|
Abstract
In recent years many new members of the family of TRP ion channels have been identified. These channels are classified into several subgroups and participate in many sensory and physiological functions. TRPV channels are important for the perception of pain, temperature sensing, osmotic regulation, and maintenance of calcium homeostasis, and much recent research concerns the identification of protein domains involved in mediating specific channel functions. Recent literature on TRPV channel subunit composition, protein domains required for subunit assembly, trafficking, and regulation will be reviewed and discussed.
Collapse
Affiliation(s)
- Barbara A Niemeyer
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Gebäude 46, 66421, Homburg, Germany.
| |
Collapse
|
29
|
Dobrydneva Y, Abelt CJ, Dovel B, Thadigiri CM, Williams RL, Blackmore PF. 2-Aminoethoxydiphenyl Borate as a Prototype Drug for a Group of Structurally Related Calcium Channel Blockers in Human Platelets. Mol Pharmacol 2005; 69:247-56. [PMID: 16214957 DOI: 10.1124/mol.105.015701] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have synthesized a series of 2-aminoethoxydiphenyl borate (2-APB, 2,2-diphenyl-1,3,2-oxazaborolidine) analogs and tested their ability to inhibit thrombin-induced Ca(2+) influx in human platelets. The analogs were either synthesized by adding various substituents to the oxazaborolidine ring (methyl, dimethyl, tert-butyl, phenyl, methyl phenyl, and pyridyl) or increasing the size of the oxazaborolidine ring to seven- and nine-membered rings. NMR analysis of the boron-containing analogs suggests that each of them exist as a ring structure through the formation of an N-->B coordinate bond (except for the hexyl analog). The possibility that these boron-containing compounds formed dimers was also considered. All compounds dose-dependently inhibited thrombin-induced Ca(2+) influx in human platelets, with the 2,2-diphenyl-1,3,2-oxazaborolidine-5-one derivative having the weakest activity at 100 microM, whereas the (S)-4-benzyl and (R)-4-benzyl derivatives of 2-APB were approximately 10 times more potent than the parent 2-APB. Two nonboron analogs (3-methyl and 3-tert-butyl 2,2-diphenyl-1,3-oxazolidine) were synthesized; they had approximately the same activity as 2-APB, and this implies that the presence of boron was not necessary for inhibitory activity. All of the compounds tested were also able to inhibit thrombin-induced calcium release. We concluded that extensive modifications of the oxazaborolidine ring in 2-APB can be made, and Ca(2+)-blocking activity was maintained.
Collapse
Affiliation(s)
- Yuliya Dobrydneva
- Department of Physiological Sciences, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23501, USA
| | | | | | | | | | | |
Collapse
|
30
|
Uehara K. Localization of TRPC1 channel in the sinus endothelial cells of rat spleen. Histochem Cell Biol 2005; 123:347-56. [PMID: 15856275 DOI: 10.1007/s00418-004-0741-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2004] [Indexed: 01/21/2023]
Abstract
The ultrastructural localization of transient receptor potential C1 (TRPC1) channels in the sinus endothelial cells of rat spleen was examined by confocal laser scanning and electron microscopy. In addition, the localization of the closely associated proteins and channels, VE-cadherin, calreticulin, inositol-1,4,5-trisphosphate receptors type 1 (IP3R1), and ryanodine receptor (RyR), was also examined. Immunofluorescence microscopy of tissue cryosections revealed TRPC1 channels to be localized within the cytoplasm, in the superficial layer of the apical and basal parts of the cells, and in the junctional area of the adjacent endothelial cells. The distribution of Ca2+-storing tubulovesicular structures within endothelial cells was established by using tissue sections treated with osmium ferricyanide. Electron microscopy revealed densely stained tubulovesicular structures closely apposed to the plasma membrane and that occasionally ran closely parallel to the plasma membrane and near the caveolae and junctional apparatus. Immunolocalization analysis at the electron microscopy level using immunogold bound to the secondary antibody confirmed that TRPC1 channels were localized in the plasma membrane, caveolae, and vesicular structures in the subplasmalemmal cytoplasm of sinus endothelial cells. Calreticulin was predominantly localized in endoplasmic reticulum. IP3R1 and RyR, considered to be type 3, were colocalized in endoplasmic reticulum in proximity to the plasma membrane and caveolae. Thus, TRPC1 channels in sinus endothelial cells of the spleen might play an important role in controlling blood cell passage through phenomena including cytoskeletal reorganization, cell retraction, and disassembly of adherens junctions.
Collapse
Affiliation(s)
- Kiyoko Uehara
- Department of Cell Biology, Fukuoka University School of Medicine, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
31
|
Rosado JA, Redondo PC, Sage SO, Pariente JA, Salido GM. Store-operated Ca2+ entry: Vesicle fusion or reversible trafficking and de novo conformational coupling? J Cell Physiol 2005; 205:262-9. [PMID: 15880447 DOI: 10.1002/jcp.20399] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Store-operated Ca2+ entry (SOCE), a mechanism regulated by the filling state of the intracellular Ca2+ stores, is a major pathway for Ca2+ influx. Hypotheses to explain the communication between the Ca2+ stores and plasma membrane (PM) have considered both the existence of small messenger molecules, such as a Ca2+-influx factor (CIF), and both stable and de novo conformational coupling between proteins in the Ca2+ store and PM. Alternatively, a secretion-like coupling model based on vesicle fusion and channel insertion in the PM has been proposed, which shares some properties with the de novo conformational coupling model, such as the role of the actin cytoskeleton and soluble N-ethylmaleimide (NEM)-sensitive-factor attachment proteins receptor (SNARE) proteins. Here we review recent progress made in the characterization of the de novo conformational coupling and the secretion-like coupling models for SOCE. We pay particular attention into the involvement of SNARE proteins and the actin cytoskeleton in both SOCE models. SNAREs are recognized as proteins involved in exocytosis, participating in vesicle transport, membrane docking, and fusion. As with secretion, a role for the cortical actin network in Ca2+ entry has been demonstrated in a number of cell types. In resting cells, the cytoskeleton may prevent the interaction between the Ca2+ stores and the PM, or preventing fusion of vesicles containing Ca2+ channels with the PM. These are processes in which SNARE proteins might play a crucial role upon cell activation by directing a precise interaction between the membrane of the transported organelle and the PM.
Collapse
Affiliation(s)
- Juan A Rosado
- Department of Physiology, University of Extremadura, Cáceres, Spain.
| | | | | | | | | |
Collapse
|
32
|
Rosado JA, Redondo PC, Salido GM, Sage SO, Pariente JA. Cleavage of SNAP-25 and VAMP-2 impairs store-operated Ca2+entry in mouse pancreatic acinar cells. Am J Physiol Cell Physiol 2005; 288:C214-21. [DOI: 10.1152/ajpcell.00241.2004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We recently reported that store-operated Ca2+entry (SOCE) in nonexcitable cells is likely to be mediated by a reversible interaction between Ca2+channels in the plasma membrane and the endoplasmic reticulum, a mechanism known as “secretion-like coupling.” As for secretion, in this model the actin cytoskeleton plays a key regulatory role. In the present study we have explored the involvement of the secretory proteins synaptosome-associated protein (SNAP-25) and vesicle-associated membrane protein (VAMP) in SOCE in pancreatic acinar cells. Cleavage of SNAP-25 and VAMPs by treatment with botulinum toxin A (BoNT A) and tetanus toxin (TeTx), respectively, effectively inhibited amylase secretion stimulated by the physiological agonist CCK-8. BoNT A significantly reduced Ca2+entry induced by store depletion using thapsigargin or CCK-8. In addition, treatment with BoNT A once SOCE had been activated reduced Ca2+influx, indicating that SNAP-25 is needed for both the activation and maintenance of SOCE in pancreatic acinar cells. VAMP-2 and VAMP-3 are expressed in mouse pancreatic acinar cells. Both proteins associate with the cytoskeleton upon Ca2+store depletion, although only VAMP-2 seems to be sensitive to TeTx. Treatment of pancreatic acinar cells with TeTx reduced the activation of SOCE without affecting its maintenance. These findings support a role for SNAP-25 and VAMP-2 in the activation of SOCE in pancreatic acinar cells and show parallels between this process and secretion in a specialized secretory cell type.
Collapse
|