1
|
Berna-Erro A, Sanchez-Collado J, Nieto-Felipe J, Macias-Diaz A, Redondo PC, Smani T, Lopez JJ, Jardin I, Rosado JA. The Ca 2+ Sensor STIM in Human Diseases. Biomolecules 2023; 13:1284. [PMID: 37759684 PMCID: PMC10526185 DOI: 10.3390/biom13091284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/11/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
The STIM family of proteins plays a crucial role in a plethora of cellular functions through the regulation of store-operated Ca2+ entry (SOCE) and, thus, intracellular calcium homeostasis. The two members of the mammalian STIM family, STIM1 and STIM2, are transmembrane proteins that act as Ca2+ sensors in the endoplasmic reticulum (ER) and, upon Ca2+ store discharge, interact with and activate the Orai/CRACs in the plasma membrane. Dysregulation of Ca2+ signaling leads to the pathogenesis of a variety of human diseases, including neurodegenerative disorders, cardiovascular diseases, cancer, and immune disorders. Therefore, understanding the mechanisms underlying Ca2+ signaling pathways is crucial for developing therapeutic strategies targeting these diseases. This review focuses on several rare conditions associated with STIM1 mutations that lead to either gain- or loss-of-function, characterized by myopathy, hematological and immunological disorders, among others, and due to abnormal activation of CRACs. In addition, we summarize the current evidence concerning STIM2 allele duplication and deletion associated with language, intellectual, and developmental delay, recurrent pulmonary infections, microcephaly, facial dimorphism, limb anomalies, hypogonadism, and congenital heart defects.
Collapse
Affiliation(s)
- Alejandro Berna-Erro
- Department of Physiology, Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10003 Caceres, Spain; (A.B.-E.); (J.N.-F.); (A.M.-D.); (P.C.R.); (J.J.L.)
| | - Jose Sanchez-Collado
- Department of Medical Physiology and Biophysics, University of Seville, 41004 Seville, Spain; (J.S.-C.); (T.S.)
| | - Joel Nieto-Felipe
- Department of Physiology, Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10003 Caceres, Spain; (A.B.-E.); (J.N.-F.); (A.M.-D.); (P.C.R.); (J.J.L.)
| | - Alvaro Macias-Diaz
- Department of Physiology, Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10003 Caceres, Spain; (A.B.-E.); (J.N.-F.); (A.M.-D.); (P.C.R.); (J.J.L.)
| | - Pedro C. Redondo
- Department of Physiology, Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10003 Caceres, Spain; (A.B.-E.); (J.N.-F.); (A.M.-D.); (P.C.R.); (J.J.L.)
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, 41004 Seville, Spain; (J.S.-C.); (T.S.)
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio, University of Seville, Spanish National Research Council (CSIC), 41004 Seville, Spain
| | - Jose J. Lopez
- Department of Physiology, Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10003 Caceres, Spain; (A.B.-E.); (J.N.-F.); (A.M.-D.); (P.C.R.); (J.J.L.)
| | - Isaac Jardin
- Department of Physiology, Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10003 Caceres, Spain; (A.B.-E.); (J.N.-F.); (A.M.-D.); (P.C.R.); (J.J.L.)
| | - Juan A. Rosado
- Department of Physiology, Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10003 Caceres, Spain; (A.B.-E.); (J.N.-F.); (A.M.-D.); (P.C.R.); (J.J.L.)
| |
Collapse
|
2
|
Kim JH, Hwang KH, Dang BTN, Eom M, Kong ID, Gwack Y, Yu S, Gee HY, Birnbaumer L, Park KS, Cha SK. Insulin-activated store-operated Ca 2+ entry via Orai1 induces podocyte actin remodeling and causes proteinuria. Nat Commun 2021; 12:6537. [PMID: 34764278 PMCID: PMC8586150 DOI: 10.1038/s41467-021-26900-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 10/28/2021] [Indexed: 12/28/2022] Open
Abstract
Podocyte, the gatekeeper of the glomerular filtration barrier, is a primary target for growth factor and Ca2+ signaling whose perturbation leads to proteinuria. However, the effects of insulin action on store-operated Ca2+ entry (SOCE) in podocytes remain unknown. Here, we demonstrated that insulin stimulates SOCE by VAMP2-dependent Orai1 trafficking to the plasma membrane. Insulin-activated SOCE triggers actin remodeling and transepithelial albumin leakage via the Ca2+-calcineurin pathway in podocytes. Transgenic Orai1 overexpression in mice causes podocyte fusion and impaired glomerular filtration barrier. Conversely, podocyte-specific Orai1 deletion prevents insulin-stimulated SOCE, synaptopodin depletion, and proteinuria. Podocyte injury and albuminuria coincide with Orai1 upregulation at the hyperinsulinemic stage in diabetic (db/db) mice, which can be ameliorated by the suppression of Orai1-calcineurin signaling. Our results suggest that tightly balanced insulin action targeting podocyte Orai1 is critical for maintaining filter integrity, which provides novel perspectives on therapeutic strategies for proteinuric diseases, including diabetic nephropathy. Perturbations of Ca2+ signaling in podocytes may deteriorate kidney function and eventually lead to proteinuria. Here the authors show that insulin can affect the function of the calcium regulator Ora1 in podocytes, which is critical for maintaining kidney filter integrity.
Collapse
Affiliation(s)
- Ji-Hee Kim
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Kyu-Hee Hwang
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Bao T N Dang
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - In Deok Kong
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Seyoung Yu
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA.,Institute of Biomedical Research (BIOMED), School of Medical Sciences, Catholic University of Argentina, C1107AAZ, Buenos Aires, Argentina
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Seung-Kuy Cha
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea. .,Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea. .,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.
| |
Collapse
|
3
|
de Souza LB, Ong HL, Liu X, Ambudkar IS. PIP 2 and septin control STIM1/Orai1 assembly by regulating cytoskeletal remodeling via a CDC42-WASP/WAVE-ARP2/3 protein complex. Cell Calcium 2021; 99:102475. [PMID: 34601312 DOI: 10.1016/j.ceca.2021.102475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/20/2022]
Abstract
Store-operated calcium entry (SOCE) is triggered by assembly of Orai1 with STIM proteins in ER-PM junctions. Plasma membrane PIP2 as well as PIP2-binding protein, SEPT4, significantly impact Orai1-STIM1 interaction. While septins and PIP2 can organize the actin cytoskeleton, it is unclear whether the status of actin within the junctions contributes to SOCE. We report herein that actin remodeling modulates STIM1 clustering. Our findings show that a PIP2- and SEPT4-dependent mechanism involving CDC42, WASP/WAVE, and ARP2 regulates actin remodeling into a ring-like structure around STIM1 puncta. CDC42 localization in the ER-plasma membrane region is enhanced following ER-Ca2+ store depletion. PIP2 depletion or knockdown of SEPT4 attenuate the recruitment of CDC42 to the ER-PM region. Importantly, knockdown of SEPT4, or CDC42+ARP2, disrupts the organization of actin as well as STIM1 clustering. Consequently, Orai1 recruitment to STIM1 puncta, SOCE, and NFAT translocation to the nucleus are all attenuated. Ca2+ influx induced by STIM1-C terminus is not affected by CDC42 knockdown. In aggregate, our findings reveal that PIP2 and SEPT4 affect Orai1/STIM1 clustering by coordinating actin remodeling within ER-PM junctions. This dynamic reorganization of actin has an important role in regulation of SOCE and downstream Ca2+-dependent effector functions.
Collapse
Affiliation(s)
- Lorena Brito de Souza
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA
| | - Hwei Ling Ong
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA.
| | - Xibao Liu
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA
| | - Indu S Ambudkar
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA.
| |
Collapse
|
4
|
Cantonero C, Camello PJ, Salido GM, Rosado JA, Redondo PC. TMEM97 facilitates the activation of SOCE by downregulating the association of cholesterol to Orai1 in MDA-MB-231 cells. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158906. [PMID: 33618021 DOI: 10.1016/j.bbalip.2021.158906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/02/2021] [Accepted: 02/11/2021] [Indexed: 01/03/2023]
Abstract
The expression of TMEM97, a regulator of cholesterol transport, has been reported to be enhanced in some tumour cells. We have recently shown that TMEM97 is involved in the proliferation of the breast cancer cell line MDA-MB-231, probably through changes in store-operated calcium entry (SOCE). By using silencing and overexpression of TMEM97 in MDA-MB-231 cells (two manoeuvres that either reduce or increase the calcium influx, respectively), we show enhanced cholesterol uptake in these cells as compared to the non-tumoral breast cell line, MCF10A. The enhanced cholesterol uptake in MDA-MB-231 cells was inhibited by silencing TMEM97, while overexpression of this protein increased cholesterol uptake in MCF10A cells and, therefore, indicating that this protein plays a role in the enhanced cholesterol uptake in MDA-MB-231 cancer cell line. TMEM97 silencing and overexpression resulted in an increase and decrease in the association of cholesterol to the SOCE calcium channel Orai1, respectively. Interestingly, silencing of TMEM97 in MDA-MB-231 cells significantly reduced the co-localization of Orai1 with the SOCE regulatory protein STIM1. Finally, neither silencing nor overexpression of TMEM97 altered SOCE in MDA-MB-231 cells transfected with the cholesterol insensible mutant of Orai1(Y80E). Our results reveal a novel regulatory mechanism of SOCE that relies on TMEM97 activity that courses through the reduction of the cholesterol content in the plasma membrane, and subsequently, by impairing its interaction with Orai1.
Collapse
Affiliation(s)
- C Cantonero
- Department of Physiology (Phycell group), University of Extremadura, Caceres 10003, Spain
| | - P J Camello
- Department of Physiology (FIMUL group), University of Extremadura, Caceres 10003, Spain
| | - G M Salido
- Department of Physiology (Phycell group), University of Extremadura, Caceres 10003, Spain
| | - J A Rosado
- Department of Physiology (Phycell group), University of Extremadura, Caceres 10003, Spain
| | - P C Redondo
- Department of Physiology (Phycell group), University of Extremadura, Caceres 10003, Spain.
| |
Collapse
|
5
|
Bai Y, Li B, Wang S, Jiang H, Li J, Wang W, Wang K, Qin L, Jia J. Effects of estrogen on STIM1/Orai1 in the sublingual gland of ovariectomized rats. Histol Histopathol 2020; 35:701-707. [PMID: 31916583 DOI: 10.14670/hh-18-198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Studies have shown that estrogen can protect the function of the sublingual gland, but the specific mechanism is still unclear. Besides, the STIM1/Orai1 pathway is important to secretion in the salivary gland. Here, we explore the possible effects of estrogen on sublingual gland function by observing changes of STIM1 and Orai1 levels in the sublingual glands of ovariectomized rats. METHODS 42 adult female Sprague-Dawley rats were randomly divided into three groups: SHAM, OVX, and OVX+E (n = 14 per group). Two weeks after ovariectomy, rats were treated with estrogen (β-estradiol). The expression of STIM1 and Orai1 in the sublingual gland were observed by double label-immunohistochemistry and immunofluorescence. Calcium imaging was conducted to observe changes in cellular Ca²⁺ levels. RESULTS IHC and IF showed that the levels of both STIM1 and Orai1 decreased following ovariectomy, but increased to SHAM levels after estrogen treatment. By IF, STIM1 and Orai1 exhibited perfect co-localization. Calcium imaging results showed that the Ca²⁺ in the cells decreased after ovariectomy. Estrogen intervention returned levels of these proteins and Ca²⁺ to the same as those in the control group. CONCLUSION This study demonstrates that low estrogen status significantly reduced the expression of STIM1 and Orai1 in the sublingual gland of rats, along with cellular Ca²⁺ levels. These data provide insight into the likely mechanisms underlying sublingual gland secretion dysfunction during menopause.
Collapse
Affiliation(s)
- Yun Bai
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China.,Department of Stomatology, Tianjin Hospital, Tianjin, China
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Sinan Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Hai Jiang
- Department of Anatomy and Histoembryology, School of Basic Medical Science, Peking University, Beijing, China
| | - Junlei Li
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Wenjuan Wang
- Department of Anatomy and Histoembryology, School of Basic Medical Science, Peking University, Beijing, China
| | - Ke Wang
- Department of Anatomy and Histoembryology, School of Basic Medical Science, Peking University, Beijing, China
| | - Lihua Qin
- Department of Anatomy and Histoembryology, School of Basic Medical Science, Peking University, Beijing, China.
| | - Jing Jia
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China.,Department of Stomatology, The Third Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, China.
| |
Collapse
|
6
|
Smani T, Domínguez-Rodriguez A, Callejo-García P, Rosado JA, Avila-Medina J. Phospholipase A2 as a Molecular Determinant of Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:111-31. [PMID: 27161227 DOI: 10.1007/978-3-319-26974-0_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Activation of phospholipases A2 (PLA2) leads to the generation of biologically active lipid products that can affect numerous cellular events. Ca(2+)-independent PLA2 (iPLA2), also called group VI phospholipase A2, is one of the main types forming the superfamily of PLA2. Beside of its role in phospholipid remodeling, iPLA2 has been involved in intracellular Ca(2+) homeostasis regulation. Several studies proposed iPLA2 as an essential molecular player of store operated Ca(2+) entry (SOCE) in a large number of excitable and non-excitable cells. iPLA2 activation releases lysophosphatidyl products, which were suggested as agonists of store operated calcium channels (SOCC) and other TRP channels. Herein, we will review the important role of iPLA2 on the intracellular Ca(2+) handling focusing on its role in SOCE regulation and its implication in physiological and/or pathological processes.
Collapse
Affiliation(s)
- 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.
| | - Alejandro Domínguez-Rodriguez
- 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
| | - Paula Callejo-García
- 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
| | - Javier Avila-Medina
- 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
| |
Collapse
|
7
|
Lopez E, Bermejo N, Berna-Erro A, Alonso N, Salido GM, Redondo PC, Rosado JA. Relationship between calcium mobilization and platelet α- and δ-granule secretion. A role for TRPC6 in thrombin-evoked δ-granule exocytosis. Arch Biochem Biophys 2015; 585:75-81. [PMID: 26386308 DOI: 10.1016/j.abb.2015.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 11/25/2022]
Abstract
Changes in cytosolic Ca(2+) concentration ([Ca(2+)]c) regulate granule secretion in different cell types. Thrombin activates PAR1 and PAR4 receptors and promotes release of Ca(2+) from distinct intracellular stores, which, in turn, activates store-operated Ca(2+) entry (SOCE). A crucial step during platelet function is the release of physiological agonists stored in secretory granules to the extracellular compartment during activation. We aim to study the role of Ca(2+) mobilization from the extracellular compartment or from different intracellular stores in platelet granule secretion. By using flow cytometry, we have found that α- and δ-granules are secreted in thrombin-stimulated platelets in the absence of extracellular Ca(2+), and in a concentration-dependent manner. Our findings show that thrombin-stimulated granule secretion depends on Ca(2+) mobilization from intracellular stores. Analysis of the kinetics of granule secretion reveals that platelet stimulation with thrombin results in rapid release of α-granules which precedes the secretion of δ-granules. Incubation of platelets with a specific antibody, which recognizes the extracellular amino acid sequence 573-586 of TRPC6, inhibited thrombin-evoked δ-granule exocytosis. Our results indicate that the mechanisms underlying thrombin-induced α- and δ-granule secretion show differences in dependency on Ca(2+) mobilization.
Collapse
Affiliation(s)
- E Lopez
- Department of Physiology, University of Extremadura, Phycell, 10003, Spain
| | - N Bermejo
- Department of Hematology, Hospital San Pedro de Alcantara, 10003 Cáceres, Spain
| | - A Berna-Erro
- Department of Physiology, University of Extremadura, Phycell, 10003, Spain
| | - N Alonso
- Department of Hematology, Hospital Infanta Cristina, 06006 Badajoz, Spain
| | - G M Salido
- Department of Physiology, University of Extremadura, Phycell, 10003, Spain
| | - P C Redondo
- Department of Physiology, University of Extremadura, Phycell, 10003, Spain
| | - J A Rosado
- Department of Physiology, University of Extremadura, Phycell, 10003, Spain.
| |
Collapse
|
8
|
A relay mechanism between EB1 and APC facilitate STIM1 puncta assembly at endoplasmic reticulum-plasma membrane junctions. Cell Calcium 2013; 54:246-56. [PMID: 23871111 DOI: 10.1016/j.ceca.2013.06.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 12/20/2022]
Abstract
The assembly of STIM1 protein puncta near endoplasmic reticulum-plasma membrane (ER-PM) junctions is required for optimal activation of store-operated channels (SOC). The mechanisms controlling the translocation of STIM1 puncta to ER-PM junctions remain largely unknown. In the present study, we have explored the role of the microtubule binding protein adenomatous polyposis coli (APC), on STIM1 puncta and store-operated calcium entry (SOCE). APC-depleted cells showed reduced STIM1 puncta near ER-PM junctions, instead puncta is found at the ER surrounding the cell nucleus. Reduced STIM1 puncta near ER-PM junctions in APC-depleted cells correlates with a strong inhibition of SOCE and diminished Orai whole-cell currents. Immunoprecipitation and confocal microscopy co-localization studies indicate that, upon depletion of the ER, STIM1 dissociates from EB1 and associates to APC. Deletion analysis identified an APC-binding domain in the carboxyl terminus of STIM1 (STIM1 650-685). These results together position APC as an important element in facilitating the translocation of STIM1 puncta near ER-PM junctions, which in turn is required for efficient SOCE and Orai activation upon depletion of the ER.
Collapse
|
9
|
López E, Berna-Erro A, Bermejo N, Brull JM, Martinez R, Garcia Pino G, Alvarado R, Salido GM, Rosado JA, Cubero JJ, Redondo PC. Long-term mTOR inhibitors administration evokes altered calcium homeostasis and platelet dysfunction in kidney transplant patients. J Cell Mol Med 2013; 17:636-47. [PMID: 23577651 PMCID: PMC3822816 DOI: 10.1111/jcmm.12044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 01/31/2013] [Indexed: 12/12/2022] Open
Abstract
The use of the mammal target of rapamycin (mTOR) inhibitors has been consolidated as the therapy of election for preventing graft rejection in kidney transplant patients, despite their immunosuppressive activity is less strong than anti-calcineurin agents like tacrolimus and cyclosporine A. Furthermore, as mTOR is widely expressed, rapamycin (a macrolide antibiotic produced by Streptomyces hygroscopicus) is recommended in patients presenting neoplasia due to its antiproliferative actions. Hence, we have investigated whether rapamycin presents side effects in the physiology of other cell types different from leucocytes, such as platelets. Blood samples were drawn from healthy volunteers and kidney transplant patients long-term medicated with rapamycin: sirolimus and everolimus. Platelets were either loaded with fura-2 or directly stimulated, and immunoassayed or fixed with Laemmli's buffer to perform the subsequent analysis of platelet physiology. Our results indicate that rapamycin evokes a biphasic time-dependent alteration in calcium homeostasis and function in platelets from kidney transplant patients under rapamycin regime, as demonstrated by the reduction in granule secretion observed and subsequent impairment of platelet aggregation in these patients compared with healthy volunteers. Platelet count was also reduced in these patients, thus 41% of patients presented thrombocytopenia. All together our results show that long-term administration of rapamycin to kidney transplant patients evokes alteration in platelet function.
Collapse
Affiliation(s)
- Esther López
- Cell Physiology Research Group, Department of Physiology, University of Extremadura, Cáceres, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
López E, Berna-Erro A, Salido GM, Rosado JA, Redondo PC. FKBP52 is involved in the regulation of SOCE channels in the human platelets and MEG 01 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:652-62. [PMID: 23228564 DOI: 10.1016/j.bbamcr.2012.11.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 11/17/2012] [Accepted: 11/30/2012] [Indexed: 12/17/2022]
Abstract
Immunophilins are FK506-binding proteins that have been involved in the regulation of calcium homeostasis, either by modulating Ca(2+) channels located in the plasma membrane or in the rough endoplasmic reticulum (RE). We have investigated whether immunophilins would participate in the regulation of stored-operated Ca(2+) entry (SOCE) in human platelets and MEG 01. Both cell types were loaded with fura-2 for determining cytosolic calcium concentration changes ([Ca(2+)](c)), or stimulated and fixed to evaluate the protein interaction profile by performing immunoprecipitation and western blotting. We have found that incubation of platelets with FK506 increases Ca(2+) mobilization. Thapsigargin (TG)-evoked, Thr-evoked SOCE and TG-evoked Mn(2+) entry resulted in significant reduction by treatment of platelets with immunophilin antagonists. We confirmed by immunoprecipitation that immunophilins interact with transient receptor potential channel 1 (TRPC1) and Orai1 in human platelets. FK506 and rapamycin reduced the association between TRPC1 and Orai1 with FK506 binding protein (52) (FKBP52) in human platelets, and between TRPC1 and the type II IP(3)R, which association is known to be crucial for the maintenance of SOCE in human platelets. FKBP52 role in SOCE activation was confirmed by silencing FKBP52 using SiRNA FKBP52 in MEG 01 as demonstrated by single cell configuration imaging technique. TRPC1 silencing and depletion of cell of TRPC1 and FKBP52 simultaneously, impair activation of SOCE evoked by TG in MEG 01. Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. All together, these results demonstrate that immunophilins participate in the regulation of SOCE in human platelets.
Collapse
Affiliation(s)
- Esther López
- Department of Physiology Cellular Physiology Research Group, University of Extremadura, 10003 Cáceres, Spain
| | | | | | | | | |
Collapse
|
11
|
Lopez E, Jardin I, Berna-Erro A, Bermejo N, Salido GM, Sage SO, Rosado JA, Redondo PC. STIM1 tyrosine-phosphorylation is required for STIM1-Orai1 association in human platelets. Cell Signal 2012; 24:1315-22. [PMID: 22387225 DOI: 10.1016/j.cellsig.2012.02.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 02/16/2012] [Accepted: 02/16/2012] [Indexed: 10/28/2022]
Abstract
Stromal interaction molecule 1 (STIM1) is a key element of the store-operated Ca(2+) entry mechanism (SOCE). Recently, regulation of STIM1 by glycosylation and phosphorylation on serine/threonine or proline residues has been described; however other modes of phosphorylation that are important for activating SOCE in platelets, such as tyrosine phosphorylation, have been poorly investigated. Here we investigate the latency of STIM1 phosphorylation on tyrosine residues during the first steps of SOCE activation. Human platelets were stimulated and fixed at desired times using rapid kinetic assays instruments, and immunoprecipitation and western blotting techniques were then used to investigate the pattern of STIM1 tyrosine phosphorylation during the first steps of SOCE activation. We have found that maximal STIM1 tyrosine phosphorylation occurred 2.5s after stimulation of human platelets with thapsigargin (Tg). STIM1 localized in the plasma membrane were also phosphorylated in platelets stimulated with Tg. By using chemical inhibitors that target different members of the Src family of tyrosine kinases (SKFs), two independent signaling pathways involved in STIM1 tyrosine phosphorylation during the first steps of SOCE activation were identified. We finally conclude that STIM1 tyrosine phosphorylation is a key event for the association of STIM1 with plasma membrane Ca(2+) channels such as Orai1, hence it is required for conducting SOCE activation.
Collapse
Affiliation(s)
- Esther Lopez
- Cell Physiology Research Group, Department of Physiology, University of Extremadura, 10003 Cáceres, Spain
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Wang L, Zhao Y, Gui B, Fu R, Ma F, Yu J, Qu P, Dong L, Chen C. Acute stimulation of glucagon secretion by linoleic acid results from GPR40 activation and [Ca2+]i increase in pancreatic islet {alpha}-cells. J Endocrinol 2011; 210:173-9. [PMID: 21565851 DOI: 10.1530/joe-11-0132] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The role of free fatty acids (FFAs) in glucagon secretion has not been well established, and the involvement of FFA receptor GPR40 and its downstream signaling pathways in regulating glucagon secretion are rarely demonstrated. In this study, it was found that linoleic acid (LA) acutely stimulated glucagon secretion from primary cultured rat pancreatic islets. LA at 20 and 40 μmol/l dose-dependently increased glucagon secretion both at 3 mmol/l glucose and at 15 mmol/l glucose, although 15 mmol/l glucose reduced basal glucagon levels. LA induced an increase in cytoplasmic free calcium concentrations ([Ca(2)(+)](i)) in identified rat α-cells, which is reflected by increased Fluo-3 intensity under confocal microscopy recording. The increase in [Ca(2)(+)](i) was partly inhibited by removal of extracellular Ca(2)(+) and eliminated overall by further exhaustion of intracellular Ca(2)(+) stores using thapsigargin treatment, suggesting that both Ca(2)(+) release and Ca(2)(+) influx contributed to the LA-stimulated increase in [Ca(2)(+)](i) in α-cells. Double immunocytochemical stainings showed that GPR40 was expressed in glucagon-positive α-cells. LA-stimulated increase in [Ca(2)(+)](i) was blocked by inhibition of GPR40 expression in α-cells after GPR40-specific antisense treatment. The inhibition of phospholipase C activity by U73122 also blocked the increase in [Ca(2)(+)](i) by LA. It is concluded that LA activates GPR40 and phospholipase C (and downstream signaling pathways) to increase Ca(2)(+) release and associated Ca(2)(+) influx through Ca(2)(+) channels, resulting in increase in [Ca(2)(+)](i) and glucagon secretion.
Collapse
Affiliation(s)
- Li Wang
- The Second Affiliated Hospital of Medical School, Xi'an Jiao Tong University, Xi'an 710004, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Valenzuela JI, Jaureguiberry-Bravo M, Couve A. Neuronal protein trafficking: emerging consequences of endoplasmic reticulum dynamics. Mol Cell Neurosci 2011; 48:269-77. [PMID: 21782949 DOI: 10.1016/j.mcn.2011.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 06/23/2011] [Accepted: 07/07/2011] [Indexed: 01/16/2023] Open
Abstract
The highly polarized morphology and complex geometry of neurons is determined to a great extent by the structural and functional organization of the secretory pathway. It is intuitive to propose that the spatial arrangement of secretory organelles and their dynamic behavior impinge on protein trafficking and neuronal function, but these phenomena and their consequences are not well delineated. Here we analyze the architecture and motility of the archetypal endoplasmic reticulum (ER), and their relationship to the microtubule cytoskeleton and post-translational modifications of tubulin. We also review the dynamics of the ER in axons, dendrites and spines, and discuss the role of ER dynamics on protein mobility and trafficking in neurons.
Collapse
Affiliation(s)
- José Ignacio Valenzuela
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | | |
Collapse
|
14
|
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
|
15
|
Jardín I, López JJ, Redondo PC, Salido GM, Rosado JA. Store-operated Ca2+ entry is sensitive to the extracellular Ca2+ concentration through plasma membrane STIM1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1614-22. [PMID: 19631699 DOI: 10.1016/j.bbamcr.2009.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 07/03/2009] [Accepted: 07/13/2009] [Indexed: 01/06/2023]
Abstract
Store-operated Ca(2+) entry (SOCE) is a major mechanism for Ca(2+) influx in platelets and other cells activated by a reduction in Ca(2+) concentration in the intracellular stores. SOCE has been reported to be regulated by extracellular Ca(2+), although the underlying mechanism remains unclear. Here we have examined the involvement of plasma membrane-located STIM1 (PM-STIM1) in the regulation of SOCE by extracellular Ca(2+). Treatment of platelets with the SERCA inhibitor thapsigargin (TG) induced Mn(2+) entry, which was inhibited by extracellular Ca(2+) in a concentration-dependent manner. Incubation of platelets with a specific antibody, which recognizes the extracellular amino acid sequence 25-139 of PM-STIM1 that contains the Ca(2+)-binding domain, prevented the inactivation of Ca(2+) entry induced by extracellular Ca(2+). TG induced translocation of STIM1 to the plasma membrane (PM), an event that was found to be Ca(2+)-dependent. In addition, TG stimulated association of PM-STIM1 with Orai1, an event that was not prevented by stabilization of the membrane cytoskeleton using jasplakinolide. These findings suggest that PM-STIM1 is important for the inactivation of SOCE by extracellular Ca(2+), an event that is likely to be mediated by interaction with Orai1.
Collapse
Affiliation(s)
- Isaac Jardín
- Department of Physiology (Cellular Physiology Research Group), University of Extremadura, 10071 Cáceres, Spain
| | | | | | | | | |
Collapse
|
16
|
Dionisio N, Garcia-Mediavilla MV, Sanchez-Campos S, Majano PL, Benedicto I, Rosado JA, Salido GM, Gonzalez-Gallego J. Hepatitis C virus NS5A and core proteins induce oxidative stress-mediated calcium signalling alterations in hepatocytes. J Hepatol 2009; 50:872-82. [PMID: 19303156 DOI: 10.1016/j.jhep.2008.12.026] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Revised: 11/18/2008] [Accepted: 12/09/2008] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIMS The hepatitis C virus (HCV) structural core and non-structural NS5A proteins induce in liver cells a series of intracellular events, including elevation of reactive oxygen and nitrogen species (ROS/RNS). Since oxidative stress is associated to altered intracellular Ca(2+) homeostasis, we aimed to investigate the effect of these proteins on Ca(2+) mobilization in human hepatocyte-derived transfected cells, and the protective effect of quercetin treatment. METHODS Ca(2+) mobilization and actin reorganization were determined by spectrofluorimetry. Production of ROS/RNS was determined by flow cytometry. RESULTS Cells transfected with NS5A and core proteins showed enhanced ROS/RNS production and resting cytosolic Ca(2+) concentration, and reduced Ca(2+) concentration into the stores. Phenylephrine-evoked Ca(2+) release, Ca(2+) entry and extrusion by the plasma membrane Ca(2+)-ATPase were significantly reduced in transfected cells. Similar effects were observed in cytokine-activated cells. Phenylephrine-evoked actin reorganization was reduced in the presence of core and NS5A proteins. These effects were significantly prevented by quercetin. Altered Ca(2+) mobilization and increased calpain activation were observed in replicon-containing cells. CONCLUSIONS NS5A and core proteins induce oxidative stress-mediated Ca(2+) homeostasis alterations in human hepatocyte-derived cells, which might underlie the effects of both proteins in the pathogenesis of liver disorders associated to HCV infection.
Collapse
Affiliation(s)
- Natalia Dionisio
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Cáceres, Spain
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Cioffi DL, Lowe K, Alvarez DF, Barry C, Stevens T. TRPing on the lung endothelium: calcium channels that regulate barrier function. Antioxid Redox Signal 2009; 11:765-76. [PMID: 18783312 PMCID: PMC2850299 DOI: 10.1089/ars.2008.2221] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rises in cytosolic calcium are sufficient to initiate the retraction of endothelial cell borders and to increase macromolecular permeability. Although endothelial cell biologists have recognized the importance of shifts in cytosolic calcium for several decades, only recently have we gained a rudimentary understanding of the membrane calcium channels that change cell shape. Members of the transient receptor potential family (TRP) are chief among the molecular candidates for permeability-coupled calcium channels. Activation of calcium entry through store-operated calcium entry channels, most notably TRPC1 and TRPC4, increases lung endothelial cell permeability, as does activation of calcium entry through the TRPV4 channel. However, TRPC1 and TRPC4 channels appear to influence the lung extraalveolar endothelial barrier most prominently, whereas TRPV4 channels appear to influence the lung capillary endothelial barrier most prominently. Thus, phenotypic heterogeneity in ion channel expression and function exists within the lung endothelium, along the arterial-capillary-venous axis, and is coupled to discrete control of endothelial barrier function.
Collapse
Affiliation(s)
- Donna L Cioffi
- Center for Lung Biology, University of South Alabama, Mobile, Alabama 36688, USA
| | | | | | | | | |
Collapse
|
18
|
Alexandru N, Jardín I, Popov D, Simionescu M, García-Estañ J, Salido GM, Rosado JA. Effect of homocysteine on calcium mobilization and platelet function in type 2 diabetes mellitus. J Cell Mol Med 2009; 12:2586-97. [PMID: 18088391 PMCID: PMC3828875 DOI: 10.1111/j.1582-4934.2008.00195.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Type 2 diabetes mellitus induces a characteristic platelet hyperactivity that might be due to several factors including oxidative stress and abnormal intracellular Ca2+ homeostasis. Hyperhomocysteinaemia is considered a risk factor in the development of thrombosis although its effect on platelet function and the mechanisms involved are still poorly understood. Here we show that homocysteine (Hcy) induce a concentration-dependent increase in endogenous production of reactive oxygen species (ROS), which was significantly greater in platelets from diabetic patients than in controls. Platelet treatment with Hcy resulted in Ca2+ release from the dense tubular system and the acidic stores. Ca2+ mobilisation-induced by Hcy consisted in two components, an initial slow increase in intracellular free Ca2+ concentration ([Ca2+]i) and a rapid and marked increase in [Ca2+]i, the second leading to the activation of platelet aggregation. As well as ROS generation, Ca2+ mobilization and platelet aggregation were significantly greater in platelets from diabetic donors than in controls, which indicate that platelets from diabetic donors are more sensitive to Hcy. These findings, together with the hyperhomocysteinaemia reported in diabetic patients, strongly suggest that Hcy might be considered a risk factor in the development of cardiovascular complications associated to type 2 diabetes mellitus.
Collapse
Affiliation(s)
- N Alexandru
- Institute of Cellular Biology and Pathology N. Simionescu, Bucharest, Romania
| | | | | | | | | | | | | |
Collapse
|
19
|
Alexandru N, Jardín I, Popov D, Simionescu M, García-Estañ J, Salido GM, Rosado JA. Effect of homocysteine on calcium mobilization and platelet function in type 2 diabetes mellitus. J Cell Mol Med 2009; 12:2015-26. [PMID: 19012728 PMCID: PMC4506167 DOI: 10.1111/j.1582-4934.2008.00200.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Type 2 diabetes mellitus induces a characteristic platelet hyperactivity that might be due to several factors including oxidativ stress and abnormal intracellular Ca2+ homeostasis. Hyperhomocysteinaemia is considered a risk factor in the development of thrombosis although its effect on platelet function and the mechanisms involved are still poorly understood. Here we show tha homocysteine induce a concentration-dependent increase in endogenous production of reactive oxygen species (ROS), which was significantly greater in platelets from diabetic patients than in controls. Platelet treatment with homocysteine resulted in Ca2+ release from the dense tubular system and the acidic stores. Ca2+ mobilization-induced by homocysteine consisted in two components, an initial slow increase in intracellular free Ca + concentration ([Ca +]i) and a rapid and marked increase in [Ca2+]i, th second leading to the activation of platelet aggregation. As well as ROS generation, Ca2+ mobilization and platelet aggregation were significantly greater in platelets from diabetic donors than in controls, which indicate that platelets from diabetic donors are more sensitive to homocysteine. These findings, together with the hyperhomocysteinaemia reported in diabetic patients, strongly suggest that homocysteine might be considered a risk factor in the development of cardiovascular complications associated to type 2 diabetes mellitus.
Collapse
Affiliation(s)
- N Alexandru
- Institute of Cellular Biology and Pathology N. Simionescu, Bucharest, Romania
| | | | | | | | | | | | | |
Collapse
|
20
|
Phenolphthalein as a prototype drug for a group of structurally related calcium channel blockers in human platelets. J Cardiovasc Pharmacol 2009; 53:231-40. [PMID: 19247192 DOI: 10.1097/fjc.0b013e31819b5494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Thrombin increases intracellular free Ca ([Ca]i) in human platelets by 2 mechanisms: internal mobilization and the influx of Ca via store-operated Ca entry (SOCE). 2-Aminoethoxydiphenyl borate (2-APB) is an inhibitor of SOCE. In search for nonboron analogues of 2-APB, we identified a well-known compound, phenolphthalein, structurally related to 2-APB. Many phenolphthalein analogues inhibited the ability of thrombin and thapsigargin (a specific activator of SOCE) to increase [Ca]i. Phenolphthalein has an IC50 approximately 10 microM to inhibit thrombin-induced [Ca]i elevation, its active analogues have a similar potency. Several phenolphthalein analogues also inhibited thrombin-induced intracellular Ca mobilization, which indicates action on inositol 1,4,5-trisphosphate receptors. We identified structural features among active and inactive phenolphthalein analogues that are responsible for the activity. Opening of the 5-membered lactone ring of phenolphthalein resulted in a total loss of activity. If the diphenyl rings possessed primary amine, dimethyl amine, or a cyano group, there was no activity. Modifications to the diphenyl groups that were tolerated include phosphate, sulfate, iodine, bromine, methyl, nitrite, and methoxy. Inhibition of thrombin-induced [Ca]i increase by phenolphthalein was not mediated by an increase in cyclic adenosine monophosphate because the inhibitor of cyclic adenosine monophosphate-dependent protein kinase A, 4-cyano-3-methylisoquinoline, did not affect the inhibitory action of phenolphthalein. The inhibitory effect of phenolphthalein was not mediated by an increase in NO/cyclic guanosine monophosphate (cGMP) because the inhibitors of NO-sensitive soluble guanylyl cyclase, methylene blue, and ODQ did not affect the inhibition. Phytohemagglutinin and thapsigargin-induced SOCE in Jurkat cells was also inhibited by phenolphthalein and 2-APB to the same extent as seen in platelets. Therefore, phenolphthalein and its derivatives structurally similar to 2-APB inhibit SOCE in platelets and other cells.
Collapse
|
21
|
Morgado S, Granados MP, Bejarano I, López JJ, Salido GM, González A, Pariente JA. Role of intracellular calcium on hydrogen peroxide-induced apoptosis in rat pancreatic acinar AR42J cells. J Appl Biomed 2008. [DOI: 10.32725/jab.2008.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
22
|
Grosse J, Braun A, Varga-Szabo D, Beyersdorf N, Schneider B, Zeitlmann L, Hanke P, Schropp P, Mühlstedt S, Zorn C, Huber M, Schmittwolf C, Jagla W, Yu P, Kerkau T, Schulze H, Nehls M, Nieswandt B. An EF hand mutation in Stim1 causes premature platelet activation and bleeding in mice. J Clin Invest 2008; 117:3540-50. [PMID: 17965774 DOI: 10.1172/jci32312] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 08/15/2007] [Indexed: 12/31/2022] Open
Abstract
Changes in cytoplasmic Ca2+ levels regulate a variety of fundamental cellular functions in virtually all cells. In nonexcitable cells, a major pathway of Ca2+ entry involves receptor-mediated depletion of intracellular Ca2+ stores followed by the activation of store-operated calcium channels in the plasma membrane. We have established a mouse line expressing an activating EF hand motif mutant of stromal interaction molecule 1 (Stim1), an ER receptor recently identified as the Ca2+ sensor responsible for activation of Ca2+ release-activated (CRAC) channels in T cells, whose function in mammalian physiology is not well understood. Mice expressing mutant Stim1 had macrothrombocytopenia and an associated bleeding disorder. Basal intracellular Ca2+ levels were increased in platelets, which resulted in a preactivation state, a selective unresponsiveness to immunoreceptor tyrosine activation motif-coupled agonists, and increased platelet consumption. In contrast, basal Ca2+ levels, but not receptor-mediated responses, were affected in mutant T cells. These findings identify Stim1 as a central regulator of platelet function and suggest a cell type-specific activation or composition of the CRAC complex.
Collapse
|
23
|
Zhang J, Blackmore PF, Hargrave BY, Xiao S, Beebe SJ, Schoenbach KH. Nanosecond pulse electric field (nanopulse): A novel non-ligand agonist for platelet activation. Arch Biochem Biophys 2008; 471:240-8. [DOI: 10.1016/j.abb.2007.12.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 12/14/2007] [Accepted: 12/15/2007] [Indexed: 11/29/2022]
|
24
|
Abstract
The anti-estrogenic drug tamoxifen, which is used therapeutically for treatment and prevention of breast cancer, can lead to the development of thrombosis. We found that tamoxifen rapidly increased intracellular free calcium [Ca2+]i in human platelets from both male and female donors. Thus 10 microM tamoxifen increased [Ca2+]i above the resting level by 197 +/- 19%. Tamoxifen acted synergistically with thrombin, ADP, and vasopressin to increase [Ca2+]i. The anti-estrogen ICI 182780 did not attenuate the effects of tamoxifen to increase [Ca2+]i; however, phospholipase C inhibitor U-73122 blocked this effect. 4-hydroxytamoxifen, a major metabolite of tamoxifen, also increased [Ca2+]i, but other tamoxifen metabolites and synthetic derivatives did not. Three hydroxylated derivatives of triphenylethylene (corresponding to the hydrophobic core of tamoxifen) which are transitional structures between tamoxifen (Ca agonist) and diethylstilbestrol (Ca antagonist) increased [Ca2+]i slightly (6% to 24%) and partially inhibited thrombin-induced [Ca2+]i elevation (68% to 79%). Therefore the dimethylaminoethyl moiety is responsible for tamoxifen being a Ca agonist rather than antagonist. 4-Hydroxytamoxifen and polymer-conjugated derivatives of 4-hydroxytamoxifen increased [Ca2+]i, with similar efficacy. The ability of tamoxifen to increase [Ca2+]i in platelets, leading to platelet activation, and its ability to act synergistically with other platelet agonists may contribute to development of tamoxifen-induced thrombosis.
Collapse
|
25
|
Intracellular Calcium Release from Human Platelets: Different Messengers for Multiple Stores. Trends Cardiovasc Med 2008; 18:57-61. [DOI: 10.1016/j.tcm.2007.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Revised: 11/22/2007] [Accepted: 12/06/2007] [Indexed: 11/24/2022]
|
26
|
Redondo PC, Salido GM, Pariente JA, Sage SO, Rosado JA. SERCA2b and 3 play a regulatory role in store-operated calcium entry in human platelets. Cell Signal 2008; 20:337-46. [DOI: 10.1016/j.cellsig.2007.10.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Accepted: 10/18/2007] [Indexed: 11/26/2022]
|
27
|
Natriuretic peptides in vascular physiology and pathology. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 268:59-93. [PMID: 18703404 DOI: 10.1016/s1937-6448(08)00803-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Four major natriuretic peptides have been isolated: atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), C-type natriuretic peptide (CNP), and Dendroaspis-type natriuretic peptide (DNP). Natriuretic peptides play an important role in the regulation of cardiovascular homeostasis maintaining blood pressure and extracellular fluid volume. The classical endocrine effects of natriuretic peptides to modulate fluid and electrolyte balance and vascular smooth muscle tone are complemented by autocrine and paracrine actions that include regulation of coronary blood flow and, therefore, myocardial perfusion; modulation of proliferative responses during myocardial and vascular remodeling; and cytoprotective anti-ischemic effects. The actions of natriuretic peptides are mediated by the specific binding of these peptides to three cell surface receptors: type A natriuretic peptide receptor (NPR-A), type B natriuretic peptide receptor (NPR-B), and type C natriuretic peptide receptor (NPR-C). NPR-A and NPR-B are guanylyl cyclase receptors that increase intracellular cGMP concentration and activate cGMP-dependent protein kinases. NPR-C has been presented as a clearance receptor and its activation also results in inhibition of adenylyl cyclase activity. The wide range of effects of natriuretic peptides might be the base for the development of new therapeutic strategies of great benefit in patients with cardiovascular problems including coronary artery disease or heart failure. This review summarizes current literature concerning natriuretic peptides, their receptors and their effects on fluid/electrolyte balance, and vascular and cardiac physiology and pathology, including primary hypertension and myocardial infarction. In addition, we will attempt to provide an update on important issues regarding natriuretic peptides in congestive heart failure.
Collapse
|
28
|
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
|
29
|
Redondo PC, Harper AGS, Harper MT, Brownlow SL, Rosado JA, Sage SO. hTRPC1-associated alpha-actinin, and not hTRPC1 itself, is tyrosine phosphorylated during human platelet activation. J Thromb Haemost 2007; 5:2476-83. [PMID: 17892531 DOI: 10.1111/j.1538-7836.2007.02773.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Canonical transient receptor potential channels (TRPCs), which are regulated by several processes, including tyrosine phosphorylation, are candidates for the conduction of store-operated Ca(2+) entry (SOCE). OBJECTIVES To assess hTRPC phosphotyrosine content upon platelet stimulation. METHODS A new protein complex immunological separation assay (ProCISA) was developed to allow assessment of isolated hTRPC tyrosine phosphorylation by Western blotting. RESULTS Classical immunoprecipitation suggested that thrombin (Thr) evoked an initial decrease in hTRPC1 phosphotyrosine content, which reached a minimum at 1 s, and then increased again, exceeding basal levels after 3 min. However, TRPC isolation from protein complexes using ProCISA revealed that hTRPC1, 4 and 5 were not tyrosine phosphorylated at rest or after Thr stimulation. Stimulation with Thr for 3 min increased the phosphotyrosine content of alpha-actinin, which shows similar electrophoretic properties to hTRPCs and coimmunoprecipitates with hTRPC1. Thr-evoked alpha-actinin tyrosine phosphorylation was increased by inhibiting the alpha-actinin phosphatase, SHP-1, which enhanced phosphorylation of the TRPC complex and SOCE. Inhibition of tyrosine phosphorylation impaired the interaction between hTRPC1 and the intracellular Ca(2+) sensor STIM1. CONCLUSIONS hTRPC1, 4 and 5 are not tyrosine phosphorylated during SOCE in human platelets although tyrosine phosphorylation is important for SOCE. The results obtained using ProCISA caution the use of classical immunoprecipitation for the determination of the tyrosine phosphorylation state of a given protein, where the presence of other proteins with similar electrophoretic mobilities may give misleading results.
Collapse
Affiliation(s)
- P C Redondo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
30
|
Moccia F. Latrunculin A depolarizes starfish oocytes. Comp Biochem Physiol A Mol Integr Physiol 2007; 148:845-52. [PMID: 17897856 DOI: 10.1016/j.cbpa.2007.08.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 08/09/2007] [Accepted: 08/23/2007] [Indexed: 11/28/2022]
Abstract
Depolymerization of the actin cytoskeleton may liberate Ca2+ from InsP3-sensitive stores in some cell types, including starfish oocytes, while inhibiting Ca2+ influx in others. However, no information is available on the modulation of membrane potential (V(m)) by actin. The present study was aimed to ascertain whether the widely employed actin depolymerizing drug, latrunculin A (Lat A), affects V(m) in mature oocytes of the starfish Astropecten aranciacus. Lat A induced a membrane depolarization which was mimicked by cytochalasin D, another popular actin disruptor, and prevented by jasplakinolide, a stabilizer of the actin network. Lat A-elicited depolarization consisted in a positive shift in V(m) which reached the threshold of activation of voltage-gated Ca2+ channels (VGCC), thus triggering an action potential. Lat A-promoted depolarization lacked the action potential in Ca2+-free sea water, while it was abolished upon removal of external Na+. Moreover, membrane depolarization was prevented by pre-injection of BAPTA and heparin, but not ryanodine. These data indicate that Lat A induces a membrane depolarization by releasing Ca2+ from InsP3Rs. The Ca2+ signal in turn activates a Ca2+-dependent Na+ entry, which causes the positive shift in V(m) and stimulates the VGCC.
Collapse
Affiliation(s)
- F Moccia
- Department of Structural and Functional Biology, University of Naples Federico II, viale Cinthia, 80126 Naples, Italy.
| |
Collapse
|
31
|
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
|
32
|
Ambudkar IS, Ong HL, Liu X, Bandyopadhyay BC, Bandyopadhyay B, Cheng KT. TRPC1: The link between functionally distinct store-operated calcium channels. Cell Calcium 2007; 42:213-23. [PMID: 17350680 DOI: 10.1016/j.ceca.2007.01.013] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 01/30/2007] [Accepted: 01/31/2007] [Indexed: 10/23/2022]
Abstract
Although store-operated calcium entry (SOCE) was identified more that two decades ago, understanding the molecular mechanisms that regulate and mediate this process continue to pose a major challenge to investigators in this field. Thus, there has been major focus on determining which of the models proposed for this mechanism is valid and conclusively establishing the components of the store-operated calcium (SOC) channel(s). The transient receptor potential canonical (TRPC) proteins have been suggested as candidate components of the elusive store-operated Ca(2+) entry channel. While all TRPCs are activated in response to agonist-stimulated phosphatidylinositol 4,5, bisphosphate (PIP(2)) hydrolysis, only some display store-dependent regulation. TRPC1 is currently the strongest candidate component of SOC and is shown to contribute to SOCE in many cell types. Heteromeric interactions of TRPC1 with other TRPCs generate diverse SOC channels. Recent studies have revealed novel components of SOCE, namely the stromal interacting molecule (STIM) and Orai proteins. While STIM1 has been suggested to be the ER-Ca(2+) sensor protein relaying the signal to the plasma membrane for activation of SOCE, Orai1 is reported to be the pore-forming component of CRAC channel that mediates SOCE in T-lymphocytes and other hematopoetic cells. Several studies now demonstrate that TRPC1 also associates with STIM1 suggesting that SOC and CRAC channels are regulated by similar molecular components. Interestingly, TRPC1 is also associated with Orai1 and a TRPC1-Orai1-STIM1 ternary complex contributes to SOC channel function. This review will focus on the diverse SOC channels formed by TRPC1 and the suggestion that TRPC1 might serve as a molecular link that determines their regulation by store-depletion.
Collapse
Affiliation(s)
- Indu S Ambudkar
- Secretory Physiology Section, GTTB, NIDCR, NIH, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Jardín I, Redondo PC, Salido GM, Rosado JA. Phosphatidylinositol 4,5-bisphosphate enhances store-operated calcium entry through hTRPC6 channel in human platelets. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1783:84-97. [PMID: 17719101 DOI: 10.1016/j.bbamcr.2007.07.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 06/29/2007] [Accepted: 07/19/2007] [Indexed: 10/23/2022]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2) is a versatile regulator of TRP channels. We report that inclusion of a PIP2 analogue, PIP2 1,2-dioctanoyl, does not induce non-capacitative Ca2+ entry per se but enhanced Ca2+ entry stimulated either by thrombin or by selective depletion of the Ca2+ stores in platelets, the dense tubular system, using 10 nM TG, and the acidic stores, using 20 microM 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ). Reduction of PIP2 levels by blocking PIP2 resynthesis with Li+ or introducing a monoclonal anti-PIP2 antibody, or sequestering PIP2 using poly-lysine, attenuated Ca2+ entry induced by thrombin, TG and TBHQ, and reduced thrombin-evoked, but not TG- or TBHQ-induced, Ca2+ release from the stores. Incubation with the anti-hTRPC1 antibody did not alter the stimulation of Ca2+ entry by PIP2, whilst introduction of anti-hTRPC6 antibody directed towards the C-terminus of hTRPC6 reduced Ca2+ and Mn2+ entry induced by thrombin, TG or TBHQ, and abolished the stimulation of Ca2+ entry by PIP2. The anti-hTRPC6 antibody, but not the anti-hTRPC1 antibody or PIP2, reduced non-capacitative Ca2+ entry by the DAG analogue 1-oleoyl-2-acetyl-sn-glycerol. In summary, hTRPC6 plays a role both in store-operated and in non-capacitative Ca2+ entry. PIP2 enhances store-operated Ca2+ entry in human platelets, most probably by stimulation of hTRPC6 channels.
Collapse
Affiliation(s)
- Isaac Jardín
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10071 Cáceres, Spain
| | | | | | | |
Collapse
|
34
|
Mueller P, Quintana A, Griesemer D, Hoth M, Pieters J. Disruption of the cortical actin cytoskeleton does not affect store operated Ca2+ channels in human T-cells. FEBS Lett 2007; 581:3557-62. [PMID: 17624329 DOI: 10.1016/j.febslet.2007.06.068] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 06/24/2007] [Accepted: 06/25/2007] [Indexed: 11/17/2022]
Abstract
Lymphocyte signaling and activation leads to the influx of extracellular Ca(2+) via the activation of Ca(2+) release activated Ca(2+) (CRAC) channels in the plasma membrane. Activation of CRAC channels occurs following emptying of the endoplasmic reticulum intracellular Ca(2+) stores. One model to explain the coupling of store-emptying to CRAC activation is the secretion-like conformational coupling model. This model proposes that store depletion increases junctions between the endoplasmic reticulum and the plasma membrane in a manner that could be regulated by the cortical actin cytoskeleton. Here, we show that stabilization or depolymerization of the actin cytoskeleton failed to affect CRAC activation. We therefore conclude that rearrangement of the actin cytoskeleton is dispensable for store-operated Ca(2+) entry in T-cells.
Collapse
Affiliation(s)
- Philipp Mueller
- Biozentrum, University of Basel, Klingelbergstrasse 70, CH 4056 Basel, Switzerland
| | | | | | | | | |
Collapse
|
35
|
Redondo PC, Harper AGS, Sage SO, Rosado JA. Dual role of tubulin-cytoskeleton in store-operated calcium entry in human platelets. Cell Signal 2007; 19:2147-54. [PMID: 17681754 DOI: 10.1016/j.cellsig.2007.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Accepted: 06/15/2007] [Indexed: 11/30/2022]
Abstract
Two mechanisms for store-operated Ca(2+) entry (SOCE) regulated by two independent Ca(2+) stores, the dense tubular system (DTS) and the acidic stores, have been described in platelets. We have previously suggested that coupling between the type II IP(3) receptor (IP(3)RII) and hTRPC1, involving reorganization of the actin microfilaments, play an important role in SOCE. However, the involvement of the tubulin microtubules, located beneath the plasma membrane, remains unclear. Here we show that the microtubule disrupting agent colchicine reduced Ca(2+) entry stimulated by low concentrations (0.1 U/mL) of thrombin, which activates SOCE mostly by depleting acidic Ca(2+)-store. Consistently, colchicine reduced SOCE activated by 2,5 di-(tertbutyl)-1,4-hydroquinone (TBHQ), which selectively depletes the acidic Ca(2+) stores. In contrast, colchicine enhanced SOCE mediated by depletion of the DTS, induced by high concentrations of thapsigargin (TG), which depletes both the acidic Ca(2+) stores and the DTS, the major releasable Ca(2+) store in platelets. These findings were confirmed by using Sr(2+) as a surrogate for Ca(2+) entry. Colchicine attenuated the coupling between IP(3)RII and hTRPC1 stimulated by thrombin while it enhanced that evoked by TG. Paclitaxel, which induces microtubular stabilization and polymerization, exerted the opposite effects on thrombin- and TG-evoked SOCE and coupling between IP(3)RII and hTRPC1 compared with colchicine. Neither colchicine nor paclitaxel altered the ability of platelets to extrude Ca(2+). These findings suggest that tubulin microtubules play a dual role in SOCE, acting as a barrier that prevents constitutive SOCE regulated by DTS, but also supporting SOCE mediated by the acidic Ca(2+) stores.
Collapse
Affiliation(s)
- Pedro C Redondo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, Downing Site, UK.
| | | | | | | |
Collapse
|
36
|
Abstract
The TRPC (transient receptor potential canonical) proteins are activated in response to agonist-stimulated PIP(2) (phosphatidylinositol 4,5-bisphosphate) hydrolysis and have been suggested as candidate components of the elusive SOC (store-operated calcium channel). TRPC1 is currently the strongest candidate component of SOC. Endogenous TRPC1 has been shown to contribute to SOCE (store-operated calcium entry) in several different cell types. However, the mechanisms involved in the regulation of TRPC1 and its exact physiological function have yet to be established. Studies from our laboratory and several others have demonstrated that TRPC1 is assembled in a signalling complex with key calcium signalling proteins in functionally specific plasma membrane microdomains. Furthermore, critical interactions between TRPC1 monomers as well as interactions between TRPC1 and other proteins determine the surface expression and function of TRPC1-containing channels. Recent studies have revealed novel regulators of TRPC1-containing SOCs and have demonstrated a common molecular basis for the regulation of CRAC (calcium-release-activated calcium) and SOC channels. In the present paper, we will revisit the role of TRPC1 in SOCE and discuss how studies with TRPC1 provide an experimental basis for validating the mechanism of SOCE.
Collapse
Affiliation(s)
- I S Ambudkar
- Building 10, Room 1N-113, Secretory Physiology Section, GTTB (Gene Therapy and Therapeutics Branch), NIDCR (National Institute of Dental And Craniofacial Research), NIH (National Institutes of Health), Bethesda, MD 20892, USA.
| |
Collapse
|
37
|
Chung SC, Limnander A, Kurosaki T, Weiss A, Korenbrot JI. Coupling Ca2+ store release to Icrac channel activation in B lymphocytes requires the activity of Lyn and Syk kinases. ACTA ACUST UNITED AC 2007; 177:317-28. [PMID: 17452533 PMCID: PMC2064139 DOI: 10.1083/jcb.200702050] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Activation of the B cell receptor complex in B lymphocytes causes Ca2+ release from intracellular stores, which, in turn, activates ion channels known as Icrac. We investigated the mechanisms that link Ca2+ store release to channel gating in DT40 B lymphocyte cell lines genetically manipulated to suppress the expression of several tyrosine kinases: Btk, Lyn, Syk, and the Blnk adaptor molecule. The simultaneous but not the independent suppression of Lyn and Syk expression prevents the activation of Icrac without interfering with thapsigargin-sensitive Ca2+ store release. Icrac activation by Ca2+ is reversed in mutant cells by the homologous expression of the missing kinases. Pharmacological inhibition of kinase activity by LavendustinA and PP2 cause the same functional deficit as the genetic suppression of enzyme expression. Biochemical assays demonstrate that kinase activity is required as a tonic signal: targets must be phosphorylated to link Ca2+ store release to Icrac gating. The action of kinases on Icrac activation does not arise from control of the expression level of the stromal interaction molecule 1 and Orai1 proteins.
Collapse
Affiliation(s)
- S Clare Chung
- Department of Physiology, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
38
|
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: 52] [Impact Index Per Article: 3.1] [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
|
39
|
Abstract
Capacitative Ca2+ entry links the emptying of intracellular Ca2+ stores to the activation of store-operated Ca2+ channels in the plasma membrane. In the twenty years since the inception of the concept of capacitative Ca2+ entry, a number of activation mechanisms have been proposed, and there has been considerable interest in the possibility that TRP channels function as store-operated channels. However, in the past two years, two major players in both the signaling and permeation mechanisms for store-operated channels have been discovered: Stim1 and the Orai proteins. Stim1 is an endoplasmic reticulum Ca2+ sensor. It appears to act by redistributing within a small component of the endoplasmic reticulum, approaching the plasma membrane, but does not seem to translocate into the plasma membrane. Stim1 signals to plasma membrane Orai proteins, which constitute pore-forming subunits of store-operated channels.
Collapse
Affiliation(s)
- James W Putney
- National Institute of Environmental Health Sciences - NIH, PO Box 12233, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
40
|
Jardin I, Ben Amor N, Bartegi A, Pariente J, Salido G, Rosado J. Differential involvement of thrombin receptors in Ca2+ release from two different intracellular stores in human platelets. Biochem J 2007; 401:167-74. [PMID: 16939417 PMCID: PMC1698687 DOI: 10.1042/bj20060888] [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] [Indexed: 01/15/2023]
Abstract
Physiological agonists increase cytosolic free Ca2+ concentration to regulate a number of cellular processes. The platelet thrombin receptors, PAR (protease-activated receptor) 1 PAR-4 and GPIb-IX-V (glycoprotein Ib-IX-V) have been described as potential contributors of thrombin-induced platelet aggregation. Platelets present two separate Ca2+ stores, the DTS (dense tubular system) and acidic organelles, differentiated by the distinct sensitivity of their respective SERCAs (sarcoplasmic/endoplasmic-reticulum Ca2+-ATPases) to TG (thapsigargin) and TBHQ [2,5-di-(tert-butyl)-1,4-hydroquinone]. However, the involvement of the thrombin receptors in Ca2+ release from each Ca2+ store remains unknown. Depletion of the DTS using ADP, which releases Ca2+ solely from the DTS, in combination with 10 nM TG, to selectively inhibit SERCA2 located on the DTS reduced Ca2+ release evoked by the PAR-1 agonist, SFLLRN, and the PAR-4 agonist, AYPGKF, by 80 and 50% respectively. Desensitization of PAR-1 and PAR-4 or pre-treatment with the PAR-1 and PAR-4 antagonists SCH 79797 and tcY-NH2 reduced Ca2+ mobilization induced by thrombin, and depletion of the DTS after desensitization or blockade of PAR-1 and PAR-4 had no significant effect on Ca2+ release stimulated by thrombin through the GPIb-IX-V receptor. Converse experiments showed that depletion of the acidic stores using TBHQ reduced Ca2+ release evoked by SFLLRN or AYPGKF, by 20 and 50% respectively, and abolished thrombin-stimulated Ca2+ release through the GPIb-IX-V receptor when PAR-1 and PAR-4 had been desensitized or blocked. Our results indicate that thrombin-induced activation of PAR-1 and PAR-4 evokes Ca2+ release from both Ca2+ stores, while activation of GPIb-IX-V by thrombin releases Ca2+ solely from the acidic compartments in human platelets.
Collapse
Affiliation(s)
- Isaac Jardin
- *Cell Physiology Research Group, Department of Physiology, University of Extremadura, 10071 Cáceres, Spain
| | - Nidhal Ben Amor
- †Unité de Recherche de Biochimie, Inst. Superieur de Biotechnologie, Monastir, Tunisia
| | - Ahgleb Bartegi
- †Unité de Recherche de Biochimie, Inst. Superieur de Biotechnologie, Monastir, Tunisia
| | - José A. Pariente
- *Cell Physiology Research Group, Department of Physiology, University of Extremadura, 10071 Cáceres, Spain
| | - Ginés M. Salido
- *Cell Physiology Research Group, Department of Physiology, University of Extremadura, 10071 Cáceres, Spain
| | - Juan A. Rosado
- *Cell Physiology Research Group, Department of Physiology, University of Extremadura, 10071 Cáceres, Spain
- To whom correspondence should be addressed (email )
| |
Collapse
|
41
|
Wu MM, Buchanan J, Luik RM, Lewis RS. Ca2+ store depletion causes STIM1 to accumulate in ER regions closely associated with the plasma membrane. ACTA ACUST UNITED AC 2006; 174:803-13. [PMID: 16966422 PMCID: PMC2064335 DOI: 10.1083/jcb.200604014] [Citation(s) in RCA: 631] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stromal interacting molecule 1 (STIM1), reported to be an endoplasmic reticulum (ER) Ca2+ sensor controlling store-operated Ca2+ entry, redistributes from a diffuse ER localization into puncta at the cell periphery after store depletion. STIM1 redistribution is proposed to be necessary for Ca2+ release–activated Ca2+ (CRAC) channel activation, but it is unclear whether redistribution is rapid enough to play a causal role. Furthermore, the location of STIM1 puncta is uncertain, with recent reports supporting retention in the ER as well as insertion into the plasma membrane (PM). Using total internal reflection fluorescence (TIRF) microscopy and patch-clamp recording from single Jurkat cells, we show that STIM1 puncta form several seconds before CRAC channels open, supporting a causal role in channel activation. Fluorescence quenching and electron microscopy analysis reveal that puncta correspond to STIM1 accumulation in discrete subregions of junctional ER located 10–25 nm from the PM, without detectable insertion of STIM1 into the PM. Roughly one third of these ER–PM contacts form in response to store depletion. These studies identify an ER structure underlying store-operated Ca2+ entry, whose extreme proximity to the PM may enable STIM1 to interact with CRAC channels or associated proteins.
Collapse
Affiliation(s)
- Minnie M Wu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
42
|
Luik RM, Wu MM, Buchanan J, Lewis RS. The elementary unit of store-operated Ca2+ entry: local activation of CRAC channels by STIM1 at ER-plasma membrane junctions. ACTA ACUST UNITED AC 2006; 174:815-25. [PMID: 16966423 PMCID: PMC2064336 DOI: 10.1083/jcb.200604015] [Citation(s) in RCA: 511] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The activation of store-operated Ca2+ entry by Ca2+ store depletion has long been hypothesized to occur via local interactions of the endoplasmic reticulum (ER) and plasma membrane, but the structure involved has never been identified. Store depletion causes the ER Ca2+ sensor stromal interacting molecule 1 (STIM1) to form puncta by accumulating in junctional ER located 10–25 nm from the plasma membrane (see Wu et al. on p. 803 of this issue). We have combined total internal reflection fluorescence (TIRF) microscopy and patch-clamp recording to localize STIM1 and sites of Ca2+ influx through open Ca2+ release–activated Ca2+ (CRAC) channels in Jurkat T cells after store depletion. CRAC channels open only in the immediate vicinity of STIM1 puncta, restricting Ca2+ entry to discrete sites comprising a small fraction of the cell surface. Orai1, an essential component of the CRAC channel, colocalizes with STIM1 after store depletion, providing a physical basis for the local activation of Ca2+ influx. These studies reveal for the first time that STIM1 and Orai1 move in a coordinated fashion to form closely apposed clusters in the ER and plasma membranes, thereby creating the elementary unit of store-operated Ca2+ entry.
Collapse
Affiliation(s)
- Riina M Luik
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
43
|
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
|
44
|
Chen Q, Pan HL. Regulation of synaptic input to hypothalamic presympathetic neurons by GABA(B) receptors. Neuroscience 2006; 142:595-606. [PMID: 16887273 DOI: 10.1016/j.neuroscience.2006.06.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Revised: 06/21/2006] [Accepted: 06/22/2006] [Indexed: 02/02/2023]
Abstract
The hypothalamic paraventricular (PVN) neurons projecting to the spinal cord and brainstem play an important role in the control of homeostasis and the sympathetic nervous system. Although GABA(B) receptors are present in the PVN, their function in the control of synaptic inputs to PVN presympathetic neurons is not clear. Using retrograde tracing and whole-cell patch-clamp recordings in rat brain slices, we determined the role of presynaptic GABA(B) receptors in regulation of glutamatergic and GABAergic inputs to spinally projecting PVN neurons. The GABA(B) receptor agonist baclofen (1-50 microM) dose-dependently decreased the frequency but not the amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) and inhibitory postsynaptic currents (sIPSCs). The effect of baclofen on sEPSCs and sIPSCs was completely blocked by 10 microM CGP52432, a selective GABA(B) receptor antagonist. Baclofen also significantly reduced the frequency of both miniature excitatory and miniature inhibitory postsynaptic currents (mEPSCs and mIPSCs). Furthermore, uncoupling pertussis toxin-sensitive G(i/o) proteins with N-ethylmaleimide abolished baclofen-induced inhibition of mEPSCs and mIPSCs. However, the inhibitory effect of baclofen on the frequency of mIPSCs and mEPSCs persisted in the presence of either Cd2+, a voltage-gated Ca2+ channel blocker, or 4-aminopyridine, a blocker of voltage-gated K+ channels. Our results suggest that activation of presynaptic GABA(B) receptors inhibits synaptic GABA and glutamate release to PVN presympathetic neurons. This presynaptic action of GABA(B) receptors is mediated by the N-ethylmaleimide-sensitive G(i/o) proteins, but independent of voltage-gated Ca2+ and K+ channels.
Collapse
Affiliation(s)
- Q Chen
- Department of Anesthesiology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | |
Collapse
|
45
|
Kuwahara M, Kuwahara M. Store-mediated calcium entry in pleural mesothelial cells. Eur J Pharmacol 2006; 542:16-21. [PMID: 16824512 DOI: 10.1016/j.ejphar.2006.05.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Revised: 05/15/2006] [Accepted: 05/16/2006] [Indexed: 11/18/2022]
Abstract
Store-mediated Ca2+ entry is thought as the main pathway for Ca2+ influx in non-excitable cells. Although a role for the actin cytoskeleton in store-mediated Ca2+ entry has been proposed in some cell types, the role of actin cytoskeleton in store-mediated Ca2+ entry is still a controversy. To address this question, the effects of cytoskeletal modifiers on store-mediated Ca2+ entry in pleural mesothelial cells were examined. Thapsigargin (1 microM) induced a sufficient signal for the activation of store-mediated Ca2+ entry in pleural mesothelial cells. In the absence of extracellular Ca2+, thapsigargin induced only a transient elevation of [Ca2+]i. Moreover, re-addition of Ca2+ increased the elevation of [Ca2+]i. Passive elevations in [Ca2+]i without thapsigargin, which is induced from Ca2+ containing solution switch to Ca2+ free solution and re-add Ca2+ containing solution, were not observed in pleural mesothelial cells. Thapsigargin-induced Ca2+ entry was still present after nifedipine (1 microM) treatment. However, SKF96365 (1 microM) blocked thapsigargin-induced Ca2+ entry. Mycalolide B (1 microM) completely disrupts actin cytoskeleton in pleural mesothelial cells, but thapsigargin-induced store-mediated Ca2+ entry was preserved. Jasplakinolide (3 microM) prevented thapsigargin-induced store-mediated Ca2+ entry. These results suggest that store-mediated Ca2+ entry in pleural mesothelial cells may be mediated by a recently proposed secretion-like coupling model for store-mediated Ca2+ entry.
Collapse
Affiliation(s)
- Masayoshi Kuwahara
- Department of Comparative Pathophysiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | | |
Collapse
|
46
|
López JJ, Salido GM, Pariente JA, Rosado JA. Interaction of STIM1 with endogenously expressed human canonical TRP1 upon depletion of intracellular Ca2+ stores. J Biol Chem 2006; 281:28254-64. [PMID: 16870612 DOI: 10.1074/jbc.m604272200] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
STIM1 (stromal interaction molecule 1) has recently been proposed to communicate the intracellular Ca(2+) stores with the plasma membrane to mediate store-operated Ca(2+) entry. Here we describe for the first time that Ca(2+) store depletion stimulates rapid STIM1 surface expression and association with endogenously expressed human canonical TRP1 (hTRPC1) independently of rises in cytosolic free Ca(2+) concentration. These events require the support of the actin cytoskeleton in human platelets, as reported for the coupling between type II inositol 1,4,5-trisphosphate receptor in the Ca(2+) stores and hTRPC1 in the plasma membrane, which has been suggested to underlie the activation of store-operated Ca(2+) entry in these cells. Electrotransjection of cells with anti-STIM1 antibody, directed toward the N-terminal sequence that includes the Ca(2+)-binding region, prevented the migration of STIM1 toward the plasma membrane, the interaction between STIM1 and hTRPC1, the coupling between hTRPC1 and type II inositol 1,4,5-trisphosphate receptor, and reduced store-operated Ca(2+) entry. These findings provide evidence for a role of STIM1 in the activation of store-operated Ca(2+) entry probably acting as a Ca(2+) sensor.
Collapse
Affiliation(s)
- José J López
- Department of Physiology, Cellular Physiology Research Group, University of Extremadura, 10071 Cáceres, Spain
| | | | | | | |
Collapse
|
47
|
Rosado JA, Nuñez AM, Lopez JJ, Pariente JA, Salido GM. Intracellular Ca2+ homeostasis and aggregation in platelets are impaired by ethanol through the generation of H2O2 and oxidation of sulphydryl groups. Arch Biochem Biophys 2006; 452:9-16. [PMID: 16824477 DOI: 10.1016/j.abb.2006.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 05/09/2006] [Accepted: 05/27/2006] [Indexed: 11/29/2022]
Abstract
The mechanisms involved in the effect of ethanol on Ca2+ entry and aggregability have been investigated in human platelets in order to shed new light on the pathogenesis of alcohol consumption. Ethanol (50 mM) induced H2O2 production in platelets by Ca2+-dependent and independent mechanisms. Ca2+ entry induced by ethanol was impaired by catalase. Ethanol reduced SOCE mediated by depletion of the 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ)-sensitive acidic stores but enhances SOCE regulated by the dense tubular system. This effect was abolished by treatment with catalase or the sulphydryl group reducing agent dithiotreitol (DTT). Similarly, the anti-aggregant effect of ethanol was prevented by platelet treatment with catalase or DTT. In conclusion we provide considerable evidence that ethanol alters Ca2+ entry and reduces thrombin-induced aggregation as a result of the generation of H2O2 and the oxidation of sulphydryl groups in human platelets.
Collapse
Affiliation(s)
- Juan A Rosado
- Department of Physiology (Cellular Physiology Research Group), University of Extremadura, 10071 Cáceres, Spain
| | | | | | | | | |
Collapse
|
48
|
Rosado JA, Núñez AM, Pariente JA, Salido GM. Alterations in intracellular calcium homeostasis and platelet aggregation induced by ethanol. Biochem Biophys Res Commun 2006; 341:917-24. [PMID: 16455046 DOI: 10.1016/j.bbrc.2006.01.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 01/16/2006] [Indexed: 11/16/2022]
Abstract
The in vitro effects of ethanol on intracellular Ca(2+) homeostasis and tyrosine phosphorylation have been investigated in human platelets in order to clarify the cellular mechanisms underlying its described anti-aggregant effects. Ethanol (1-50 mM) reduced, in a dose-dependent manner, the rate and amplitude of aggregation and attenuated the phosphotyrosine content both induced by 0.1U/ml of the physiological ligand, thrombin. Thrombin-induced Ca(2+) entry to the cytosol was significantly reduced, and capacitative Ca(2+) entry (CCE) significantly altered, by 50 mM ethanol, so that ethanol reduces CCE mediated by depletion of the 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ)-sensitive store but enhances CCE induced by the TBHQ-insensitive pool. In conclusion, we provide considerable evidence that ethanol reduces thrombin-induced aggregation, which is likely a result of a significant inhibition of Ca(2+) entry, as well as a reduction in the activity of protein tyrosine kinases.
Collapse
Affiliation(s)
- Juan A Rosado
- Department of Physiology, University of Extremadura, 10071 Cáceres, Spain.
| | | | | | | |
Collapse
|
49
|
Redondo PC, Harper MT, Rosado JA, Sage SO. A role for cofilin in the activation of store-operated calcium entry by de novo conformational coupling in human platelets. Blood 2006; 107:973-9. [PMID: 16234361 DOI: 10.1182/blood-2005-05-2015] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractStore-operated Ca2+ entry (SOCE) is a major mechanism for Ca2+ influx in platelets and other cells. De novo conformational coupling between elements in the plasma membrane and Ca2+ stores, where the actin cytoskeleton plays an important regulatory role, has been proposed as the most likely mechanism to activate SOCE in platelets. Here we have examined for the first time changes in platelet F-actin levels on a subsecond time scale. Using stopped-flow fluorimetry and a quenched-flow approach, we provide evidence for the involvement of cofilin in actin filament reorganization and SOCE in platelets. Thrombin (0.1 U/mL) evoked an initial decrease in F-actin that commenced within 0.1 second and reached a minimum 0.9 second after stimulation, prior to the activation of SOCE. F-actin then increased, exceeding basal levels approximately 2.5 seconds after stimulation. Thrombin also induced cofilin dephosphorylation and activation, which paralleled the changes observed in F-actin, and rapid Btk activation. Inhibition of cofilin dephosphorylation by LFM-A13 resulted in the loss of net actin depolymerization and an increased delay in SOCE initiation. These results suggest that cofilin is important for the rapid actin remodeling necessary for the activation of SOCE in platelets through de novo conformational coupling.
Collapse
Affiliation(s)
- Pedro C Redondo
- Department of Physiology, University of Extremadura, Cáceres, Spain
| | | | | | | |
Collapse
|
50
|
Rosado JA, Lopez JJ, Gomez-Arteta E, Redondo PC, Salido GM, Pariente JA. Early caspase-3 activation independent of apoptosis is required for cellular function. J Cell Physiol 2006; 209:142-52. [PMID: 16791842 DOI: 10.1002/jcp.20715] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A number of pro-apoptotic stimuli induce the activation of caspase-9, an initiator protease that activates executioner caspases, such as caspase-3, leading to the development of programmed cell death. Here we demonstrate that cell (platelets and pancreatic acinar cells) stimulation with agonists induces a bimodal activation of caspase-3. The early caspase-3 activation occurs within 1 min of stimulation and is independent on caspase-9 or mitochondrial cytochrome c release suggesting that is a non-apoptotic event. The ability of agonists to induce early activation of caspase-3 is similar to that observed for other physiological processes. Activation of caspase-3 by physiological concentrations of cellular agonists, including thrombin or CCK-8, is independent of rises in cytosolic calcium concentration but requires PKC activation, and is necessary for agonist-induced activation of the tyrosine kinases Btk and pp60src and for several cellular functions, including store-operated calcium entry, platelet aggregation, or pancreatic secretion. Thus, early activation of caspase-3 seems to be a non-apoptotic event required for cellular function.
Collapse
Affiliation(s)
- Juan A Rosado
- Department of Physiology (Cellular Physiology Research Group), University of Extremadura, Cáceres, Spain.
| | | | | | | | | | | |
Collapse
|