1
|
Badaut J, Blochet C, Obenaus A, Hirt L. Physiological and pathological roles of caveolins in the central nervous system. Trends Neurosci 2024; 47:651-664. [PMID: 38972795 PMCID: PMC11324375 DOI: 10.1016/j.tins.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 05/14/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Caveolins are a family of transmembrane proteins located in caveolae, small lipid raft invaginations of the plasma membrane. The roles of caveolin-enriched lipid rafts are diverse, and include mechano-protection, lipid homeostasis, metabolism, transport, and cell signaling. Caveolin-1 (Cav-1) and other caveolins were described in endothelial cells and later in other cell types of the central nervous system (CNS), including neurons, astrocytes, oligodendrocytes, microglia, and pericytes. This pancellular presence of caveolins demands a better understanding of their functional roles in each cell type. In this review we describe the various functions of Cav-1 in the cells of normal and pathological brains. Several emerging preclinical findings suggest that Cav-1 could represent a potential therapeutic target in brain disorders.
Collapse
Affiliation(s)
- Jérôme Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Camille Blochet
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - André Obenaus
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA; Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Lorenz Hirt
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Li W, Guo J, Hobson EC, Xue X, Li Q, Fu J, Deng CX, Guo Z. Metabolic-Glycoengineering-Enabled Molecularly Specific Acoustic Tweezing Cytometry for Targeted Mechanical Stimulation of Cell Surface Sialoglycans. Angew Chem Int Ed Engl 2024; 63:e202401921. [PMID: 38498603 PMCID: PMC11073901 DOI: 10.1002/anie.202401921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 03/20/2024]
Abstract
In this study, we developed a novel type of dibenzocyclooctyne (DBCO)-functionalized microbubbles (MBs) and validated their attachment to azide-labelled sialoglycans on human pluripotent stem cells (hPSCs) generated by metabolic glycoengineering (MGE). This enabled the application of mechanical forces to sialoglycans on hPSCs through molecularly specific acoustic tweezing cytometry (mATC), that is, displacing sialoglycan-anchored MBs using ultrasound (US). It was shown that subjected to the acoustic radiation forces of US pulses, sialoglycan-anchored MBs exhibited significantly larger displacements and faster, more complete recovery after each pulse than integrin-anchored MBs, indicating that sialoglycans are more stretchable and elastic than integrins on hPSCs in response to mechanical force. Furthermore, stimulating sialoglycans on hPSCs using mATC reduced stage-specific embryonic antigen-3 (SSEA-3) and GD3 expression but not OCT4 and SOX2 nuclear localization. Conversely, stimulating integrins decreased OCT4 nuclear localization but not SSEA-3 and GD3 expression, suggesting that mechanically stimulating sialoglycans and integrins initiated distinctive mechanoresponses during the early stages of hPSC differentiation. Taken together, these results demonstrated that MGE-enabled mATC uncovered not only different mechanical properties of sialoglycans on hPSCs and integrins but also their different mechanoregulatory impacts on hPSC differentiation, validating MGE-based mATC as a new, powerful tool for investigating the roles of glycans and other cell surface biomolecules in mechanotransduction.
Collapse
Affiliation(s)
- Weiping Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiatong Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Eric C. Hobson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qingjiang Li
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Jianping Fu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cheri X. Deng
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
3
|
Brignone MS, Lanciotti A, Molinari P, Mallozzi C, De Nuccio C, Caprini ES, Petrucci TC, Visentin S, Ambrosini E. Megalencephalic leukoencephalopathy with subcortical cysts protein-1: A new calcium-sensitive protein functionally activated by endoplasmic reticulum calcium release and calmodulin binding in astrocytes. Neurobiol Dis 2024; 190:106388. [PMID: 38141856 DOI: 10.1016/j.nbd.2023.106388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/04/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND MLC1 is a membrane protein highly expressed in brain perivascular astrocytes and whose mutations account for the rare leukodystrophy (LD) megalencephalic leukoencephalopathy with subcortical cysts disease (MLC). MLC is characterized by macrocephaly, brain edema and cysts, myelin vacuolation and astrocyte swelling which cause cognitive and motor dysfunctions and epilepsy. In cultured astrocytes, lack of functional MLC1 disturbs cell volume regulation by affecting anion channel (VRAC) currents and the consequent regulatory volume decrease (RVD) occurring in response to osmotic changes. Moreover, MLC1 represses intracellular signaling molecules (EGFR, ERK1/2, NF-kB) inducing astrocyte activation and swelling following brain insults. Nevertheless, to date, MLC1 proper function and MLC molecular pathogenesis are still elusive. We recently reported that in astrocytes MLC1 phosphorylation by the Ca2+/Calmodulin-dependent kinase II (CaMKII) in response to intracellular Ca2+ release potentiates MLC1 activation of VRAC. These results highlighted the importance of Ca2+ signaling in the regulation of MLC1 functions, prompting us to further investigate the relationships between intracellular Ca2+ and MLC1 properties. METHODS We used U251 astrocytoma cells stably expressing wild-type (WT) or mutated MLC1, primary mouse astrocytes and mouse brain tissue, and applied biochemistry, molecular biology, video imaging and electrophysiology techniques. RESULTS We revealed that WT but not mutant MLC1 oligomerization and trafficking to the astrocyte plasma membrane is favored by Ca2+ release from endoplasmic reticulum (ER) but not by capacitive Ca2+ entry in response to ER depletion. We also clarified the molecular events underlining MLC1 response to cytoplasmic Ca2+ increase, demonstrating that, following Ca2+ release, MLC1 binds the Ca2+ effector protein calmodulin (CaM) at the carboxyl terminal where a CaM binding sequence was identified. Using a CaM inhibitor and generating U251 cells expressing MLC1 with CaM binding site mutations, we found that CaM regulates MLC1 assembly, trafficking and function, being RVD and MLC-linked signaling molecules abnormally regulated in these latter cells. CONCLUSION Overall, we qualified MLC1 as a Ca2+ sensitive protein involved in the control of volume changes in response to ER Ca2+ release and astrocyte activation. These findings provide new insights for the comprehension of the molecular mechanisms responsible for the myelin degeneration occurring in MLC and other LD where astrocytes have a primary role in the pathological process.
Collapse
Affiliation(s)
- M S Brignone
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - A Lanciotti
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - P Molinari
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - C Mallozzi
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - C De Nuccio
- Istituto Superiore di Sanità, Research Coordination and Support Service, Viale Regina Elena 299, 00161 Rome, Italy
| | - E S Caprini
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - T C Petrucci
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - S Visentin
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - E Ambrosini
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
4
|
Brignone MS, Lanciotti A, Michelucci A, Mallozzi C, Camerini S, Catacuzzeno L, Sforna L, Caramia M, D’Adamo MC, Ceccarini M, Molinari P, Macioce P, Macchia G, Petrucci TC, Pessia M, Visentin S, Ambrosini E. The CaMKII/MLC1 Axis Confers Ca2+-Dependence to Volume-Regulated Anion Channels (VRAC) in Astrocytes. Cells 2022; 11:cells11172656. [PMID: 36078064 PMCID: PMC9454758 DOI: 10.3390/cells11172656] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Astrocytes, the main glial cells of the central nervous system, play a key role in brain volume control due to their intimate contacts with cerebral blood vessels and the expression of a distinctive equipment of proteins involved in solute/water transport. Among these is MLC1, a protein highly expressed in perivascular astrocytes and whose mutations cause megalencephalic leukoencephalopathy with subcortical cysts (MLC), an incurable leukodystrophy characterized by macrocephaly, chronic brain edema, cysts, myelin vacuolation, and astrocyte swelling. Although, in astrocytes, MLC1 mutations are known to affect the swelling-activated chloride currents (ICl,swell) mediated by the volume-regulated anion channel (VRAC), and the regulatory volume decrease, MLC1′s proper function is still unknown. By combining molecular, biochemical, proteomic, electrophysiological, and imaging techniques, we here show that MLC1 is a Ca2+/Calmodulin-dependent protein kinase II (CaMKII) target protein, whose phosphorylation, occurring in response to intracellular Ca2+ release, potentiates VRAC-mediated ICl,swell. Overall, these findings reveal that MLC1 is a Ca2+-regulated protein, linking volume regulation to Ca2+ signaling in astrocytes. This knowledge provides new insight into the MLC1 protein function and into the mechanisms controlling ion/water exchanges in the brain, which may help identify possible molecular targets for the treatment of MLC and other pathological conditions caused by astrocyte swelling and brain edema.
Collapse
Affiliation(s)
| | - Angela Lanciotti
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Antonio Michelucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Serena Camerini
- Core Facilities (FAST), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Martino Caramia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Maria Cristina D’Adamo
- Department of Medicine and Surgery, LUM Giuseppe Degennaro University, 70010 Bari, Italy
| | - Marina Ceccarini
- National Centre for Rare Diseases, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Paola Molinari
- National Centre for Drug Research and Evaluation (FARVA), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Pompeo Macioce
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | | | | | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Sergio Visentin
- National Centre for Drug Research and Evaluation (FARVA), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
- Correspondence: ; Tel.: +39-06-4990-2037
| |
Collapse
|
5
|
Cremer T, Neefjes J, Berlin I. The journey of Ca 2+ through the cell - pulsing through the network of ER membrane contact sites. J Cell Sci 2020; 133:133/24/jcs249136. [PMID: 33376155 DOI: 10.1242/jcs.249136] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Calcium is the third most abundant metal on earth, and the fundaments of its homeostasis date back to pre-eukaryotic life forms. In higher organisms, Ca2+ serves as a cofactor for a wide array of (enzymatic) interactions in diverse cellular contexts and constitutes the most important signaling entity in excitable cells. To enable responsive behavior, cytosolic Ca2+ concentrations are kept low through sequestration into organellar stores, particularly the endoplasmic reticulum (ER), but also mitochondria and lysosomes. Specific triggers are then used to instigate a local release of Ca2+ on demand. Here, communication between organelles comes into play, which is accomplished through intimate yet dynamic contacts, termed membrane contact sites (MCSs). The field of MCS biology in relation to cellular Ca2+ homeostasis has exploded in recent years. Taking advantage of this new wealth of knowledge, in this Review, we invite the reader on a journey of Ca2+ flux through the ER and its associated MCSs. New mechanistic insights and technological advances inform the narrative on Ca2+ acquisition and mobilization at these sites of communication between organelles, and guide the discussion of their consequences for cellular physiology.
Collapse
Affiliation(s)
- Tom Cremer
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Ilana Berlin
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| |
Collapse
|
6
|
Denizot A, Arizono M, Nägerl UV, Soula H, Berry H. Simulation of calcium signaling in fine astrocytic processes: Effect of spatial properties on spontaneous activity. PLoS Comput Biol 2019; 15:e1006795. [PMID: 31425510 PMCID: PMC6726244 DOI: 10.1371/journal.pcbi.1006795] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 09/04/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Astrocytes, a glial cell type of the central nervous system, have emerged as detectors and regulators of neuronal information processing. Astrocyte excitability resides in transient variations of free cytosolic calcium concentration over a range of temporal and spatial scales, from sub-microdomains to waves propagating throughout the cell. Despite extensive experimental approaches, it is not clear how these signals are transmitted to and integrated within an astrocyte. The localization of the main molecular actors and the geometry of the system, including the spatial organization of calcium channels IP3R, are deemed essential. However, as most calcium signals occur in astrocytic ramifications that are too fine to be resolved by conventional light microscopy, most of those spatial data are unknown and computational modeling remains the only methodology to study this issue. Here, we propose an IP3R-mediated calcium signaling model for dynamics in such small sub-cellular volumes. To account for the expected stochasticity and low copy numbers, our model is both spatially explicit and particle-based. Extensive simulations show that spontaneous calcium signals arise in the model via the interplay between excitability and stochasticity. The model reproduces the main forms of calcium signals and indicates that their frequency crucially depends on the spatial organization of the IP3R channels. Importantly, we show that two processes expressing exactly the same calcium channels can display different types of calcium signals depending on the spatial organization of the channels. Our model with realistic process volume and calcium concentrations successfully reproduces spontaneous calcium signals that we measured in calcium micro-domains with confocal microscopy and predicts that local variations of calcium indicators might contribute to the diversity of calcium signals observed in astrocytes. To our knowledge, this model is the first model suited to investigate calcium dynamics in fine astrocytic processes and to propose plausible mechanisms responsible for their variability.
Collapse
Affiliation(s)
- Audrey Denizot
- INRIA, F-69603, Villeurbanne, France
- Univ Lyon, LIRIS, UMR5205 CNRS, F-69621, Villeurbanne, France
| | - Misa Arizono
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - U. Valentin Nägerl
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - Hédi Soula
- INRIA, F-69603, Villeurbanne, France
- Univ P&M Curie, CRC, INSERM UMRS 1138, F-75006, Paris, France
| | - Hugues Berry
- INRIA, F-69603, Villeurbanne, France
- Univ Lyon, LIRIS, UMR5205 CNRS, F-69621, Villeurbanne, France
| |
Collapse
|
7
|
Bramini M, Chiacchiaretta M, Armirotti A, Rocchi A, Kale DD, Martin C, Vázquez E, Bandiera T, Ferroni S, Cesca F, Benfenati F. An Increase in Membrane Cholesterol by Graphene Oxide Disrupts Calcium Homeostasis in Primary Astrocytes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900147. [PMID: 30891923 DOI: 10.1002/smll.201900147] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/18/2019] [Indexed: 05/24/2023]
Abstract
The use of graphene nanomaterials (GNMs) for biomedical applications targeted to the central nervous system is exponentially increasing, although precise information on their effects on brain cells is lacking. In this work, the molecular changes induced in cortical astrocytes by few-layer graphene (FLG) and graphene oxide (GO) flakes are addressed. The results show that exposure to FLG/GO does not affect cell viability or proliferation. However, proteomic and lipidomic analyses unveil alterations in several cellular processes, including intracellular Ca2+ ([Ca2+ ]i ) homeostasis and cholesterol metabolism, which are particularly intense in cells exposed to GO. Indeed, GO exposure impairs spontaneous and evoked astrocyte [Ca2+ ]i signals and induces a marked increase in membrane cholesterol levels. Importantly, cholesterol depletion fully rescues [Ca2+ ]i dynamics in GO-treated cells, indicating a causal relationship between these GO-mediated effects. The results indicate that exposure to GNMs alters intracellular signaling in astrocytes and may impact astrocyte-neuron interactions.
Collapse
Affiliation(s)
- Mattia Bramini
- Center for Synaptic Neuroscience and Technology and Graphene Labs, Istituto Italiano di Tecnologia, 16132, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| | - Martina Chiacchiaretta
- Center for Synaptic Neuroscience and Technology and Graphene Labs, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab and Graphene Labs, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Anna Rocchi
- Center for Synaptic Neuroscience and Technology and Graphene Labs, Istituto Italiano di Tecnologia, 16132, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| | - Deepali D Kale
- PharmaChemistry Line and Graphene Labs, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Cristina Martin
- Departamento de Química Orgánica, Instituto Regional de Investigación Científica Aplicada (IRICA), Universidad de Castilla La-Mancha, 13071, Ciudad Real, Spain
| | - Ester Vázquez
- Departamento de Química Orgánica, Instituto Regional de Investigación Científica Aplicada (IRICA), Universidad de Castilla La-Mancha, 13071, Ciudad Real, Spain
| | - Tiziano Bandiera
- PharmaChemistry Line and Graphene Labs, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology and Graphene Labs, Istituto Italiano di Tecnologia, 16132, Genova, Italy
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology and Graphene Labs, Istituto Italiano di Tecnologia, 16132, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| |
Collapse
|
8
|
|
9
|
Fadeel B, Bussy C, Merino S, Vázquez E, Flahaut E, Mouchet F, Evariste L, Gauthier L, Koivisto AJ, Vogel U, Martín C, Delogu LG, Buerki-Thurnherr T, Wick P, Beloin-Saint-Pierre D, Hischier R, Pelin M, Candotto Carniel F, Tretiach M, Cesca F, Benfenati F, Scaini D, Ballerini L, Kostarelos K, Prato M, Bianco A. Safety Assessment of Graphene-Based Materials: Focus on Human Health and the Environment. ACS NANO 2018; 12:10582-10620. [PMID: 30387986 DOI: 10.1021/acsnano.8b04758] [Citation(s) in RCA: 313] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Graphene and its derivatives are heralded as "miracle" materials with manifold applications in different sectors of society from electronics to energy storage to medicine. The increasing exploitation of graphene-based materials (GBMs) necessitates a comprehensive evaluation of the potential impact of these materials on human health and the environment. Here, we discuss synthesis and characterization of GBMs as well as human and environmental hazard assessment of GBMs using in vitro and in vivo model systems with the aim to understand the properties that underlie the biological effects of these materials; not all GBMs are alike, and it is essential that we disentangle the structure-activity relationships for this class of materials.
Collapse
Affiliation(s)
- Bengt Fadeel
- Nanosafety & Nanomedicine Laboratory, Institute of Environmental Medicine , Karolinska Institutet , 17777 Stockholm , Sweden
| | - Cyrill Bussy
- Nanomedicine Laboratory, Faculty of Biology, Medicine & Health , University of Manchester , Manchester M13 9PL , United Kingdom
| | - Sonia Merino
- Faculty of Chemical Science and Technology , University of Castilla-La Mancha , 13071 Ciudad Real , Spain
| | - Ester Vázquez
- Faculty of Chemical Science and Technology , University of Castilla-La Mancha , 13071 Ciudad Real , Spain
| | | | | | | | - Laury Gauthier
- CNRS, Université Paul Sabatier , 31062 Toulouse , France
| | - Antti J Koivisto
- National Research Centre for the Working Environment , 2100 Copenhagen , Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment , 2100 Copenhagen , Denmark
| | - Cristina Martín
- University of Strasbourg, CNRS , Immunology, Immunopathology and Therapeutic Chemistry , 67000 Strasbourg , France
| | - Lucia G Delogu
- Department of Chemistry and Pharmacy University of Sassari , Sassari 7100 , Italy
- Istituto di Ricerca Pediatrica , Fondazione Città della Speranza , 35129 Padova , Italy
| | - Tina Buerki-Thurnherr
- Swiss Federal Laboratories for Materials Science and Technology (EMPA) , 9014 St. Gallen , Switzerland
| | - Peter Wick
- Swiss Federal Laboratories for Materials Science and Technology (EMPA) , 9014 St. Gallen , Switzerland
| | | | - Roland Hischier
- Swiss Federal Laboratories for Materials Science and Technology (EMPA) , 9014 St. Gallen , Switzerland
| | - Marco Pelin
- Department of Life Sciences , University of Trieste , 34127 Trieste , Italy
| | | | - Mauro Tretiach
- Department of Life Sciences , University of Trieste , 34127 Trieste , Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology , Istituto Italiano di Tecnologia , 16132 Genova , Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology , Istituto Italiano di Tecnologia , 16132 Genova , Italy
| | - Denis Scaini
- Scuola Internazionale Superiore di Studi Avanzati (SISSA) , 34136 Trieste , Italy
| | - Laura Ballerini
- Scuola Internazionale Superiore di Studi Avanzati (SISSA) , 34136 Trieste , Italy
| | - Kostas Kostarelos
- Nanomedicine Laboratory, Faculty of Biology, Medicine & Health , University of Manchester , Manchester M13 9PL , United Kingdom
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences , University of Trieste , 34127 Trieste , Italy
- Carbon Nanobiotechnology Laboratory , CIC BiomaGUNE , 20009 San Sebastian , Spain
- Basque Foundation for Science, Ikerbasque , 48013 Bilbao , Spain
| | - Alberto Bianco
- University of Strasbourg, CNRS , Immunology, Immunopathology and Therapeutic Chemistry , 67000 Strasbourg , France
| |
Collapse
|
10
|
Nordzieke DE, Medraño-Fernandez I. The Plasma Membrane: A Platform for Intra- and Intercellular Redox Signaling. Antioxidants (Basel) 2018; 7:antiox7110168. [PMID: 30463362 PMCID: PMC6262572 DOI: 10.3390/antiox7110168] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/15/2018] [Accepted: 11/17/2018] [Indexed: 02/06/2023] Open
Abstract
Membranes are of outmost importance to allow for specific signal transduction due to their ability to localize, amplify, and direct signals. However, due to the double-edged nature of reactive oxygen species (ROS)—toxic at high concentrations but essential signal molecules—subcellular localization of ROS-producing systems to the plasma membrane has been traditionally regarded as a protective strategy to defend cells from unwanted side-effects. Nevertheless, specialized regions, such as lipid rafts and caveolae, house and regulate the activated/inhibited states of important ROS-producing systems and concentrate redox targets, demonstrating that plasma membrane functions may go beyond acting as a securing lipid barrier. This is nicely evinced by nicotinamide adenine dinucleotide phosphate (NADPH)-oxidases (NOX), enzymes whose primary function is to generate ROS and which have been shown to reside in specific lipid compartments. In addition, membrane-inserted bidirectional H2O2-transporters modulate their conductance precisely during the passage of the molecules through the lipid bilayer, ensuring time-scaled delivery of the signal. This review aims to summarize current evidence supporting the role of the plasma membrane as an organizing center that serves as a platform for redox signal transmission, particularly NOX-driven, providing specificity at the same time that limits undesirable oxidative damage in case of malfunction. As an example of malfunction, we explore several pathological situations in which an inflammatory component is present, such as inflammatory bowel disease and neurodegenerative disorders, to illustrate how dysregulation of plasma-membrane-localized redox signaling impacts normal cell physiology.
Collapse
Affiliation(s)
- Daniela E Nordzieke
- Institute of Microbiology and Genetics, Department of Genetics of Eukaryotic Microorganisms, Georg August University Göttingen, Grisebachstr. 8, D-37077 Göttingen, Germany.
| | - Iria Medraño-Fernandez
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
11
|
Zhang B, Paffett ML, Naik JS, Jernigan NL, Walker BR, Resta TC. Cholesterol Regulation of Pulmonary Endothelial Calcium Homeostasis. CURRENT TOPICS IN MEMBRANES 2018; 82:53-91. [PMID: 30360783 DOI: 10.1016/bs.ctm.2018.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cholesterol is a key structural component and regulator of lipid raft signaling platforms critical for cell function. Such regulation may involve changes in the biophysical properties of lipid microdomains or direct protein-sterol interactions that alter the function of ion channels, receptors, enzymes, and membrane structural proteins. Recent studies have implicated abnormal membrane cholesterol levels in mediating endothelial dysfunction that is characteristic of pulmonary hypertensive disorders, including that resulting from long-term exposure to hypoxia. Endothelial dysfunction in this setting is characterized by impaired pulmonary endothelial calcium entry and an associated imbalance that favors production vasoconstrictor and mitogenic factors that contribute to pulmonary hypertension. Here we review current knowledge of cholesterol regulation of pulmonary endothelial Ca2+ homeostasis, focusing on the role of membrane cholesterol in mediating agonist-induced Ca2+ entry and its components in the normal and hypertensive pulmonary circulation.
Collapse
Affiliation(s)
- Bojun Zhang
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Michael L Paffett
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States.
| |
Collapse
|
12
|
Montes de Oca Balderas P, Montes de Oca Balderas H. Synaptic neuron-astrocyte communication is supported by an order of magnitude analysis of inositol tris-phosphate diffusion at the nanoscale in a model of peri-synaptic astrocyte projection. BMC BIOPHYSICS 2018; 11:3. [PMID: 29456837 PMCID: PMC5809920 DOI: 10.1186/s13628-018-0043-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/30/2018] [Indexed: 12/13/2022]
Abstract
Background Astrocytes were conceived for decades only as supporting cells of the brain. However, the observation of Ca2+ waves in astrocyte synctitia, their neurotransmitter receptor expression and gliotransmitter secretion suggested a role in information handling, conception that has some controversies. Synaptic Neuron-Astrocyte metabotropic communication mediated by Inositol tris-phosphate (SN-AmcIP3) is supported by different reports. However, some models contradict this idea and Ca2+ stores are 1000 ± 325 nm apart from the Postsynaptic Density in the Perisynaptic Astrocyte Projections (PAP’s), suggesting that SN-AmcIP3 is extrasynaptic. However, this assumption does not consider IP3 Diffusion Coefficient (Dab), that activates IP3 Receptor (IP3R) releasing Ca2+ from intracellular stores. Results In this work we idealized a model of a PAP (PAPm) to perform an order of magnitude analysis of IP3 diffusion using a transient mass diffusion model. This model shows that IP3 forms a concentration gradient along the PAPm that reaches the steady state in milliseconds, three orders of magnitude before IP3 degradation. The model predicts that IP3 concentration near the Ca2+ stores may activate IP3R, depending upon Phospholipase C (PLC) number and activity. Moreover, the PAPm supports that IP3 and extracellular Ca2+ entry synergize to promote global Ca2+ transients. Conclusion The model presented here indicates that Ca2+ stores position in PAP’s does not limit SN-AmcIP3. Electronic supplementary material The online version of this article (10.1186/s13628-018-0043-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pavel Montes de Oca Balderas
- Unit of Dynamic Neurobiology, Neurochemistry Deprtment Instituto Nacional de Neurología y Neurocirugía, Insurgentes Sur #3877, Col. La Fama, C.P. 14269 Ciudad de México, Mexico
| | - Horacio Montes de Oca Balderas
- Unit of Dynamic Neurobiology, Neurochemistry Deprtment Instituto Nacional de Neurología y Neurocirugía, Insurgentes Sur #3877, Col. La Fama, C.P. 14269 Ciudad de México, Mexico
| |
Collapse
|
13
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
14
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 964] [Impact Index Per Article: 137.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
15
|
Zhang B, Naik JS, Jernigan NL, Walker BR, Resta TC. Reduced membrane cholesterol after chronic hypoxia limits Orai1-mediated pulmonary endothelial Ca 2+ entry. Am J Physiol Heart Circ Physiol 2017; 314:H359-H369. [PMID: 29101179 DOI: 10.1152/ajpheart.00540.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Endothelial dysfunction in chronic hypoxia (CH)-induced pulmonary hypertension is characterized by reduced store-operated Ca2+ entry (SOCE) and diminished Ca2+-dependent production of endothelium-derived vasodilators. We recently reported that SOCE in pulmonary arterial endothelial cells (PAECs) is tightly regulated by membrane cholesterol and that decreased membrane cholesterol is responsible for impaired SOCE after CH. However, the ion channels involved in cholesterol-sensitive SOCE are unknown. We hypothesized that cholesterol facilitates SOCE in PAECs through the interaction of Orai1 and stromal interaction molecule 1 (STIM1). The role of cholesterol in Orai1-mediated SOCE was initially assessed using CH exposure in rats (4 wk, 380 mmHg) as a physiological stimulus to decrease PAEC cholesterol. The effects of Orai1 inhibition with AnCoA4 on SOCE were examined in isolated PAEC sheets from control and CH rats after cholesterol supplementation, substitution of endogenous cholesterol with epicholesterol (Epichol), or vehicle treatment. Whereas cholesterol restored endothelial SOCE in CH rats, both Epichol and AnCoA4 attenuated SOCE only in normoxic controls. The Orai1 inhibitor had no further effect in cells pretreated with Epichol. Using cultured pulmonary endothelial cells to allow better mechanistic analysis of the molecular components of cholesterol-regulated SOCE, we found that Epichol, AnCoA4, and Orai1 siRNA each inhibited SOCE compared with their respective controls. Epichol had no additional effect after knockdown of Orai1. Furthermore, Epichol substitution significantly reduced STIM1-Orai1 interactions as assessed by a proximity ligation assay. We conclude that membrane cholesterol is required for the STIM1-Orai1 interaction necessary to elicit endothelial SOCE. Furthermore, reduced PAEC membrane cholesterol after CH limits Orai1-mediated SOCE. NEW & NOTEWORTHY This research demonstrates a novel contribution of cholesterol to regulate the interaction of Orai1 and stromal interaction molecule 1 required for pulmonary endothelial store-operated Ca2+ entry. The results provide a mechanistic basis for impaired pulmonary endothelial Ca2+ influx after chronic hypoxia that may contribute to pulmonary hypertension.
Collapse
Affiliation(s)
- Bojun Zhang
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| |
Collapse
|
16
|
Karam CS, Javitch JA. Phosphorylation of the Amino Terminus of the Dopamine Transporter: Regulatory Mechanisms and Implications for Amphetamine Action. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:205-234. [PMID: 29413521 DOI: 10.1016/bs.apha.2017.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amphetamines (AMPHs) are potent psychostimulants that are widely used and abused, with profound medical and societal impact. Their actions at dopaminergic neurons are thought to mediate their therapeutic efficacy as well as their liability for abuse and dependence. AMPHs target the dopamine transporter (DAT), the plasmalemmal membrane protein that mediates the inactivation of released dopamine (DA) through its reuptake. AMPHs act as substrates for DAT and are known to cause mobilization of dopamine (DA) to the cell exterior via DAT-mediated reverse transport (efflux). It has become increasingly evident that the mechanisms that regulate AMPH-induced DA efflux are distinct from those that regulate DA uptake. Central to these mechanisms is the phosphorylation of the DAT amino (N)-terminus, which has been repeatedly demonstrated to facilitate DAT-mediated DA efflux, without impacting other aspects of DAT physiology. This review aims to summarize the current status of knowledge regarding DAT N-terminal phosphorylation and its regulation by protein modulators and the membrane microenvironment. A better understanding of these mechanisms may lead to the identification of novel therapeutic approaches that interfere selectively with the pharmacological effects of AMPHs without altering the physiological function of DAT.
Collapse
Affiliation(s)
- Caline S Karam
- College of Physicians & Surgeons, Columbia University, New York, NY, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Jonathan A Javitch
- College of Physicians & Surgeons, Columbia University, New York, NY, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States.
| |
Collapse
|
17
|
Lakk M, Yarishkin O, Baumann JM, Iuso A, Križaj D. Cholesterol regulates polymodal sensory transduction in Müller glia. Glia 2017; 65:2038-2050. [PMID: 28856727 DOI: 10.1002/glia.23213] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/21/2022]
Abstract
Over- and underexposure to cholesterol activates glia in neurodegenerative brain and retinal diseases but the molecular targets of cholesterol in glial cells are not known. Here, we report that disruption of unesterified membrane cholesterol content modulates the transduction of chemical, mechanical and temperature stimuli in mouse Müller cells. Activation of TRPV4 (transient receptor potential vanilloid type 4), a nonselective polymodal cation channel was studied following the removal or supplementation of cholesterol using the methyl-beta cyclodextrin (MβCD) delivery vehicle. Cholesterol extraction disrupted lipid rafts and caveolae without affecting TRPV4 trafficking or membrane localization protein. However, MβCD suppressed agonist (GSK1016790A)- and temperature-evoked elevations in [Ca2+ ]i , and suppressed transcellular propagation of Ca2+ waves. Lowering the free membrane cholesterol content markedly prolonged the time-course of the glial swelling response, whereas MβCD:cholesterol supplementation enhanced agonist- and temperature-induced Ca2+ signals and shortened the swelling response. Taken together, these data show that membrane cholesterol modulates polymodal transduction of agonists, swelling and temperature stimuli in retinal radial glia and suggest that dyslipidemic retinas might be associated with abnormal glial transduction of ambient sensory inputs.
Collapse
Affiliation(s)
- Monika Lakk
- Departments of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, Utah
| | - Oleg Yarishkin
- Departments of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, Utah
| | | | - Anthony Iuso
- Departments of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, Utah
| | - David Križaj
- Departments of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, Utah
- Bioengineering, University of Utah, Salt Lake City, Utah
- Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| |
Collapse
|
18
|
TRPC Channels and Neuron Development, Plasticity, and Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:95-110. [PMID: 28508316 DOI: 10.1007/978-94-024-1088-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this chapter, we mainly focus on the functions of TRPC channels in brain development, including neural progenitor proliferation, neurogenesis, neuron survival, axon guidance, dendritic morphology, synaptogenesis, and neural plasticity. We also notice emerging advances in understanding the functions of TRPC channels in periphery, especially their functions in sensation and nociception in dorsal root ganglion (DRG). Because TRPC channels are expressed in all major types of glial cells, which account for at least half of total cells in the brain, TRPC channels may act as modulators for glial functions as well. The future challenges for studying these channels could be (1) the detailed protein structures of these channels, (2) their cell type-specific functions, (3) requirement for their specific blockers or activators, and (4) change in the channel conformation in the brain.
Collapse
|
19
|
Svobodova B, Groschner K. Reprint of "Mechanisms of lipid regulation and lipid gating in TRPC channels". Cell Calcium 2016; 60:133-41. [PMID: 27431463 DOI: 10.1016/j.ceca.2016.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 01/04/2023]
Abstract
TRPC proteins form cation channels that integrate and relay cellular signals by mechanisms involving lipid recognition and lipid-dependent gating. The lipohilic/amphiphilic molecules that function as cellular activators or modulators of TRPC proteins span a wide range of chemical structures. In this context, cellular redox balance is likely linked to the lipid recognition/gating features of TRPC channels. Both classical ligand-protein interactions as well as indirect and promiscuous sensory mechanisms have been proposed. Some of the recognition processes are suggested to involve ancillary lipid-binding scaffolds or regulators as well as dynamic protein-protein interactions determined by bilayer architecture. A complex interplay of protein-protein and protein-lipid interactions is likely to govern the gating and/or plasma membrane recruitment of TRPC channels, thereby providing a distinguished platform for signal integration and coincident signal detection. Both the primary molecular event(s) of lipid recognition by TRPC channels as well as the transformation of these events into distinct gating movements is poorly understood at the molecular level, and it remains elusive whether lipid sensing in TRPCs is conferred to a distinct sensor domain. Recent structural information on the molecular action of lipophilic activators in distantly related members of the TRP superfamily encourages speculations on TRPC gating mechanisms involved in lipid recognition/gating. This review aims to provide an update on the current understanding of the lipid-dependent control of TRPC channels with focus on the TRPC lipid sensing, signal-integration hub and a short discussion of potential links to redox signaling.
Collapse
Affiliation(s)
- Barbora Svobodova
- Institute of Biophysics, Medical University of Graz, A-8010 Graz, Austria
| | - Klaus Groschner
- Institute of Biophysics, Medical University of Graz, A-8010 Graz, Austria.
| |
Collapse
|
20
|
Mechanisms of lipid regulation and lipid gating in TRPC channels. Cell Calcium 2016; 59:271-9. [PMID: 27125985 DOI: 10.1016/j.ceca.2016.03.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 12/15/2022]
Abstract
TRPC proteins form cation channels that integrate and relay cellular signals by mechanisms involving lipid recognition and lipid-dependent gating. The lipohilic/amphiphilic molecules that function as cellular activators or modulators of TRPC proteins span a wide range of chemical structures. In this context, cellular redox balance is likely linked to the lipid recognition/gating features of TRPC channels. Both classical ligand-protein interactions as well as indirect and promiscuous sensory mechanisms have been proposed. Some of the recognition processes are suggested to involve ancillary lipid-binding scaffolds or regulators as well as dynamic protein-protein interactions determined by bilayer architecture. A complex interplay of protein-protein and protein-lipid interactions is likely to govern the gating and/or plasma membrane recruitment of TRPC channels, thereby providing a distinguished platform for signal integration and coincident signal detection. Both the primary molecular event(s) of lipid recognition by TRPC channels as well as the transformation of these events into distinct gating movements is poorly understood at the molecular level, and it remains elusive whether lipid sensing in TRPCs is conferred to a distinct sensor domain. Recent structural information on the molecular action of lipophilic activators in distantly related members of the TRP superfamily encourages speculations on TRPC gating mechanisms involved in lipid recognition/gating. This review aims to provide an update on the current understanding of the lipid-dependent control of TRPC channels with focus on the TRPC lipid sensing, signal-integration hub and a short discussion of potential links to redox signaling.
Collapse
|
21
|
Naeem M, McDaid LJ, Harkin J, Wade JJ, Marsland J. On the role of astroglial syncytia in self-repairing spiking neural networks. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2015; 26:2370-80. [PMID: 25576582 DOI: 10.1109/tnnls.2014.2382334] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
It has been shown that brain-like self-repair can arise from the interactions between neurons and astrocytes where endocannabinoids are synthesized and released from active neurons. This retrograde messenger feeds back to local synapses directly and indirectly to distant synapses via astrocytes. This direct/indirect feedback of the endocannabinoid retrograde messenger results in the modulation of the probability of release (PR) at synaptic sites. When synapses fail, there is a corresponding falloff in the firing activity of the associated neurons, and hence the strength of the direct feedback messenger diminishes. This triggers an increase in PR of healthy synapses, due to the indirect messenger from other active neurons, which is the catalyst for the repair process. In this paper, the repair process is implemented by developing a new learning rule that captures the spike-timing-dependent plasticity and Bienenstock, Cooper, and Munro learning rules. The rule is activated by the increase in PR and results in a potentiation of the weight values, which reestablishes the firing activity of neurons. In addition, this self-repairing mechanism is extended to network-level repair where astrocyte to astrocyte communications are implemented using a linear gap junction model. This facilitates the implementation of an astroglial syncytium involving multiple astrocytes, which relays the indirect feedback messenger to distant neurons: each astrocyte is bidirectionally coupled to neurons. A detailed and comprehensive set of results with analysis is presented demonstrating repair at both cellular and network levels.
Collapse
|
22
|
Larson VA, Zhang Y, Bergles DE. Electrophysiological properties of NG2(+) cells: Matching physiological studies with gene expression profiles. Brain Res 2015; 1638:138-160. [PMID: 26385417 DOI: 10.1016/j.brainres.2015.09.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/31/2015] [Accepted: 09/08/2015] [Indexed: 01/11/2023]
Abstract
NG2(+) glial cells are a dynamic population of non-neuronal cells that give rise to myelinating oligodendrocytes in the central nervous system. These cells express numerous ion channels and neurotransmitter receptors, which endow them with a complex electrophysiological profile that is unique among glial cells. Despite extensive analysis of the electrophysiological properties of these cells, relatively little was known about the molecular identity of the channels and receptors that they express. The generation of new RNA-Seq datasets for NG2(+) cells has provided the means to explore how distinct genes contribute to the physiological properties of these progenitors. In this review, we systematically compare the results obtained through RNA-Seq transcriptional analysis of purified NG2(+) cells to previous physiological and molecular studies of these cells to define the complement of ion channels and neurotransmitter receptors expressed by NG2(+) cells in the mammalian brain and discuss the potential significance of the unique physiological properties of these cells. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Valerie A Larson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ye Zhang
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
23
|
Billaud M, Lohman AW, Johnstone SR, Biwer LA, Mutchler S, Isakson BE. Regulation of cellular communication by signaling microdomains in the blood vessel wall. Pharmacol Rev 2014; 66:513-69. [PMID: 24671377 DOI: 10.1124/pr.112.007351] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.
Collapse
Affiliation(s)
- Marie Billaud
- Dept. of Molecular Physiology and Biophysics, University of Virginia School of Medicine, PO Box 801394, Charlottesville, VA 22902.
| | | | | | | | | | | |
Collapse
|
24
|
Roh SE, Hong YH, Jang DC, Kim J, Kim SJ. Lipid rafts serve as signaling platforms for mGlu1 receptor-mediated calcium signaling in association with caveolin. Mol Brain 2014; 7:9. [PMID: 24512690 PMCID: PMC3937055 DOI: 10.1186/1756-6606-7-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/30/2014] [Indexed: 11/24/2022] Open
Abstract
Background Group I metabotropic glutamate receptors (mGlu1/5 receptors) have important roles in synaptic activity in the central nervous system. They modulate neuronal excitability by mobilizing intracellular Ca2+ following receptor activation. Also, accumulating evidence has indicated the association of Ca2+ signaling with lipid rafts. Caveolin, an adaptor protein found in a specialized subset of lipid rafts, has been reported to promote the localization of membrane proteins to lipid rafts. Results In the present study, we investigated the role of lipid rafts on the mGlu1α receptor-mediated Ca2+ signaling in association with caveolin in hippocampal primary neurons and HEK293 cells. We show that the disruption of lipid rafts using methyl-β-cyclodextrin markedly decreased mGlu1α receptor-mediated Ca2+ transients and lipid rafts localization of the receptor. Furthermore, transfection of mGlu1α receptor with mutated caveolin-binding domain reduced localization of the receptor to lipid rafts. Also, application of a peptide blocker of mGlu1α receptor and caveolin binding reduced the Ca2+ signaling and the lipid rafts localization. Conclusions Taken together, these results suggest that the binding of mGlu1α receptor to caveolin is crucial for its lipid rafts localization and mGlu1α receptor-mediated Ca2+ transients.
Collapse
Affiliation(s)
| | | | | | | | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, 28, Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea.
| |
Collapse
|
25
|
Zhang H, Liu Y, Xu J, Zhang F, Liang H, Du X, Zhang H. Membrane microdomain determines the specificity of receptor-mediated modulation of Kv7/M potassium currents. Neuroscience 2013; 254:70-9. [PMID: 24036375 DOI: 10.1016/j.neuroscience.2013.08.064] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 08/28/2013] [Accepted: 08/29/2013] [Indexed: 01/18/2023]
Abstract
The Kv7/M current is one of the major mechanisms controlling neuronal excitability, which can be modulated by activation of the G protein-coupled receptor (GPCR) via distinct signaling pathways. Membrane microdomains known as lipid rafts have been implicated in the specificity of various cell signaling pathways. The aim of this study was to understand the role of lipid rafts in the specificity of Kv7/M current modulation by activation of GPCR. Methyl-β-cyclodextrin (MβCD), often used to disrupt the integrity of lipid rafts, significantly reduced the bradykinin receptor (B2R)-induced but not muscarinic receptor (M1R)-induced inhibition of the Kv7/M current. B2R and related signaling molecules but not M1R were found in caveolin-containing raft fractions of the rat superior cervical ganglia. Furthermore, activation of B2R resulted in translocation of additional B2R into the lipid rafts, which was not observed for the activation of M1R. The increase of B2R-induced intracellular Ca(2+) was also greatly reduced after MβCD treatment. Finally, B2R but not M1R was found to interact with the IP3 receptor. In conclusion, the present study implicates an important role for lipid rafts in mediating specificity for GPCR-mediated inhibition of the Kv7/M current.
Collapse
Affiliation(s)
- H Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei Province, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, Hebei Province, China; Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Mound A, Rodat-Despoix L, Bougarn S, Ouadid-Ahidouch H, Matifat F. Molecular interaction and functional coupling between type 3 inositol 1,4,5-trisphosphate receptor and BKCa channel stimulate breast cancer cell proliferation. Eur J Cancer 2013; 49:3738-51. [PMID: 23992640 DOI: 10.1016/j.ejca.2013.07.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/05/2013] [Accepted: 07/11/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND The implication of ion channels and inositol 1,4,5-trisphosphate (IP3)-induced Ca(2+) signalling (IICS) in the carcinogenesis processes, including deregulation of cell proliferation, migration and invasion, is increasingly studied. Studies from our laboratory have shown that type 3 IP3 receptor (IP3R3) and voltage- and Ca(2+)-dependent K(+) channels BKCa channels are involved in human breast cancer cell proliferation. In this context, we investigated the probable interaction between these two proteins (IP3R3 and BKCa channel) in normal and in breast cancer cells. METHODS MCF-7 and MCF-10A cell viability was measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)-assay in the presence or absence of adenosine triphosphate (ATP). Furthermore, cell-cycle analysis was carried out and cell cycle protein expression was examined by Western blotting. Immunocytochemistry and co-immunoprecipitation assays were used to check co-localisation of BKCa and IP3R3 and their molecular interaction. Finally, whole cell patch-clamp and Ca(2+) imaging were performed to assess the functional interaction. RESULTS Our results are in favour of a functional and a molecular coupling between IP3R3 and BKCa channel that is involved in MCF-7 proliferation. Indeed, ATP increased MCF-7 cell proliferation and this effect was impaired when the expression of BKCa and/or IP3R3 has been reduced by specific small interfering RNAs (siRNAs). Flow cytometry experiments showed that both siRNAs led to cell cycle arrest in the G0/G1 phase and these results were confirmed by the analysis of cell cycle protein expression. Specifically, BKCa and IP3R3 silencing decreased both cyclin-D1 and cyclin-dependent kinase 4 (CDK4) expression levels. Furthermore, ATP elicited a phospholipase C (PLC)-dependent elevation of internal Ca(2+) that triggered in turn an iberiotoxin (IbTx)- and a tetra-ethyl-ammonium (TEA)-sensitive membrane hyperpolarisation that was strongly reduced in the cells with silenced IP3R3 or BKCa. In the same way, intracellular application of Ins(2,4,5)P3 triggered an IbTx-sensitive membrane hyperpolarisation. Moreover, intracellular Ca(2+) chelation with 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) prevented ATP-induced BKCa activation. BKCa and IP3R3 also co-immunoprecipitated and this interaction seemed to occur in cholesterol-enriched microdomains. Conversely, in the normal breast cell line MCF-10A, neither ATP application nor BKCa silencing affected cell proliferation. Furthermore, IP3R3 and BKCa did not co-immunoprecipitate, suggesting the absence of a molecular coupling between BKCa and IP3R3 in the MCF-10A normal cell line. CONCLUSION Altogether, our results suggest a molecular and functional link between BKCa channel and IP3R3 in cancer cells. Our findings led us to propose this coupling between BKCa and IP3R3 as an important mechanism for tumour cell proliferation.
Collapse
Affiliation(s)
- Abdallah Mound
- Laboratory of Cellular and Molecular Physiology (EA-4667), 'Ion Channels in Breast Cancer', SFR CAP-SANTE (FED-4231), University of Amiens, UFR Sciences, 33 Rue Saint-Leu, 80039 Amiens, France
| | | | | | | | | |
Collapse
|
27
|
The membrane raft protein Flotillin-1 is essential in dopamine neurons for amphetamine-induced behavior in Drosophila. Mol Psychiatry 2013; 18:824-33. [PMID: 22710269 PMCID: PMC3582826 DOI: 10.1038/mp.2012.82] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The dopamine transporter (DAT) is the primary molecular target responsible for the rewarding properties of the psychostimulants amphetamine (AMPH) and cocaine. AMPH increases extracellular dopamine (DA) by promoting its nonexocytotic release via DAT-mediated efflux. Previous studies in heterologous cells have shown that phosphorylation of the amino terminus of DAT is required for AMPH-induced DA efflux but not for DA uptake. However, the identity of many of the modulatory proteins and the molecular mechanisms that coordinate efflux and the ensuing behavioral effects remain poorly defined. Here, we establish a robust assay for AMPH-induced hyperlocomotion in Drosophila melanogaster larvae. Using a variety of genetic and pharmacological approaches, we demonstrate that this behavioral response is dependent on DA and on DAT and its phosphorylation. We also show that methylphenidate (MPH), which competitively inhibits DA uptake but does not induce DAT-mediated DA efflux, also leads to DAT-dependent hyperlocomotion, but this response is independent of DAT phosphorylation. Moreover, we demonstrate that the membrane raft protein Flotillin-1 is required for AMPH-induced, but not MPH-induced, hyperlocomotion. These results are the first evidence of a role for a raft protein in an AMPH-mediated behavior. Thus, using our assay we are able to translate molecular and cellular findings to a behavioral level and to differentiate in vivo the distinct mechanisms of two psychostimulants.
Collapse
|
28
|
Krishnan G, Chatterjee N. Detergent resistant membrane fractions are involved in calcium signaling in Müller glial cells of retina. Int J Biochem Cell Biol 2013; 45:1758-66. [PMID: 23732110 DOI: 10.1016/j.biocel.2013.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 05/20/2013] [Accepted: 05/21/2013] [Indexed: 12/23/2022]
Abstract
Compartmentalization of the plasma membrane into lipid microdomains promotes efficient cellular processes by increasing local molecular concentrations. Calcium signaling, either as transients or propagating waves require integration of complex macromolecular machinery. Calcium waves represent a form of intercellular signaling in the central nervous system and the retina. We hypothesized that the mechanism for calcium waves would require effector proteins to aggregate at the plasma membrane in lipid microdomains. The current study shows that in Müller glia of the retina, proteins involved in calcium signaling aggregate in detergent resistant membranes identifying rafts and respond by redistributing on stimulation. We have investigated Purinoreceptor-1 (P2Y1), Ryanodine receptor (RyR), and Phospholipase C (PLC-β1). P2Y1, RyR and PLC-β1, redistribute from caveolin-1 and flotillin-1 positive fractions on stimulation with the agonists, ATP, 2MeS-ATP and Thapsigargin, an inhibitor of sarcoplasmic-endoplasmic reticulum Ca-ATPase (SERCA). Redistribution is absent on treatment with cyclopiazonic acid, another SERCA inhibitor. Disruption of rafts by removing cholesterol cause proteins involved in this machinery to redistribute and change agonist-induced calcium signaling. Cholesterol depletion from raft lead to increase in time to peak of calcium levels in agonist-evoked calcium signals in all instances, as seen by live imaging. This study emphasizes the necessity of a sub-population of proteins to cluster in specialized lipid domains. The requirement for such an organization at the raft-like microdomains may have implications on intercellular communication in the retina. Such concerted interaction at the rafts can regulate calcium dynamics and could add another layer of complexity to calcium signaling in cells.
Collapse
Affiliation(s)
- Gopinath Krishnan
- Department of L & T Ocular Pathology, Vision Research Foundation, Sankara Nethralaya, Chennai 600006, India
| | | |
Collapse
|
29
|
Tovey SC, Taylor CW. Cyclic AMP directs inositol (1,4,5)-trisphosphate-evoked Ca2+ signalling to different intracellular Ca2+ stores. J Cell Sci 2013; 126:2305-13. [PMID: 23525004 DOI: 10.1242/jcs.126144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cholesterol depletion reversibly abolishes carbachol-evoked Ca(2+) release from inositol (1,4,5)-trisphosphate (IP3)-sensitive stores, without affecting the distribution of IP3 receptors (IP3R) or endoplasmic reticulum, IP3 formation or responses to photolysis of caged IP3. Receptors that stimulate cAMP formation do not alone evoke Ca(2+) signals, but they potentiate those evoked by carbachol. We show that these potentiated signals are entirely unaffected by cholesterol depletion and that, within individual cells, different IP3-sensitive Ca(2+) stores are released by carbachol alone and by carbachol combined with receptors that stimulate cAMP formation. We suggest that muscarinic acetylcholine receptors in lipid rafts deliver IP3 at high concentration to associated IP3R, stimulating them to release Ca(2+). Muscarinic receptors outside rafts are less closely associated with IP3R and provide insufficient local IP3 to activate IP3R directly. These IP3R, probably type 2 IP3R within a discrete Ca(2+) store, are activated only when their sensitivity is increased by cAMP. Sensitization of IP3R by cAMP extends the effective range of signalling by phospholipase C, allowing muscarinic receptors that are otherwise ineffective to recruit additional IP3-sensitive Ca(2+) stores.
Collapse
Affiliation(s)
- Stephen C Tovey
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | | |
Collapse
|
30
|
Al-Saif A, Bohlega S, Al-Mohanna F. Loss of ERLIN2 function leads to juvenile primary lateral sclerosis. Ann Neurol 2013; 72:510-6. [PMID: 23109145 DOI: 10.1002/ana.23641] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Primary lateral sclerosis (PLS) is a motor neuron disorder that exclusively affects upper motor neurons leading to their degeneration. Mutations in the ALS2 gene encoding the protein Alsin have been described previously in the juvenile form of the disease. In this study, we identify mutation of the ERLIN2 gene in juvenile PLS patients and describe an in vitro model for loss of ERLIN2 function. METHODS Single nucleotide polymorphism arrays were used for homozygosity mapping. DNA sequencing of candidate genes was used to detect the underlying mutation. Level of ERLIN2 mRNA was measured by quantitative real time polymerase chain reaction. Knocking down ERLIN2 in NSC34 cells was accomplished by short-hairpin RNA interference. RESULTS We identified a splice junction mutation in the ERLIN2 gene-a component of the endoplasmic reticulum (ER) lipid rafts-that resulted in abnormal splicing of ERLIN2 transcript and nonsense-mediated decay of ERLIN2 mRNA. Knocking down ERLIN2 in NSC34 cells suppressed their growth in culture. INTERPRETATION Recently, we found that mutation of SIGMAR1, a component of ER lipid rafts, leads to juvenile amyotrophic lateral sclerosis. The identification of mutation in another component of the ER lipid rafts in juvenile PLS patients emphasizes their role in motor neuron function. Furthermore, the discovered effect of ERLIN2 loss on cell growth may advance understanding of the mechanism behind motor neuron degeneration in PLS.
Collapse
Affiliation(s)
- Amr Al-Saif
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | | | | |
Collapse
|
31
|
Verkhratsky A, Reyes RC, Parpura V. TRP channels coordinate ion signalling in astroglia. Rev Physiol Biochem Pharmacol 2013; 166:1-22. [PMID: 23784619 DOI: 10.1007/112_2013_15] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Astroglial excitability is based on highly spatio-temporally coordinated fluctuations of intracellular ion concentrations, among which changes in Ca(2+) and Na(+) take the leading role. Intracellular signals mediated by Ca(2+) and Na(+) target numerous molecular cascades that control gene expression, energy production and numerous homeostatic functions of astrocytes. Initiation of Ca(2+) and Na(+) signals relies upon plasmalemmal and intracellular channels that allow fluxes of respective ions down their concentration gradients. Astrocytes express several types of TRP channels of which TRPA1 channels are linked to regulation of functional expression of GABA transporters, whereas TRPV4 channels are activated following osmotic challenges and are up-regulated in ischaemic conditions. Astrocytes also ubiquitously express several isoforms of TRPC channels of which heteromers assembled from TRPC1, 4 and/or 5 subunits that likely act as stretch-activated channels and are linked to store-operated Ca(2+) entry. The TRPC channels mediate large Na(+) fluxes that are associated with the endoplasmic reticulum Ca(2+) signalling machinery and hence coordinate Na(+) and Ca(2+) signalling in astroglia.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK,
| | | | | |
Collapse
|
32
|
Wade J, McDaid L, Harkin J, Crunelli V, Kelso S. Self-repair in a bidirectionally coupled astrocyte-neuron (AN) system based on retrograde signaling. Front Comput Neurosci 2012; 6:76. [PMID: 23055965 PMCID: PMC3458420 DOI: 10.3389/fncom.2012.00076] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 09/10/2012] [Indexed: 11/17/2022] Open
Abstract
In this paper we demonstrate that retrograde signaling via astrocytes may underpin self-repair in the brain. Faults manifest themselves in silent or near silent neurons caused by low transmission probability (PR) synapses; the enhancement of the transmission PR of a healthy neighboring synapse by retrograde signaling can enhance the transmission PR of the "faulty" synapse (repair). Our model of self-repair is based on recent research showing that retrograde signaling via astrocytes can increase the PR of neurotransmitter release at damaged or low transmission PR synapses. The model demonstrates that astrocytes are capable of bidirectional communication with neurons which leads to modulation of synaptic activity, and that indirect signaling through retrograde messengers such as endocannabinoids leads to modulation of synaptic transmission PR. Although our model operates at the level of cells, it provides a new research direction on brain-like self-repair which can be extended to networks of astrocytes and neurons. It also provides a biologically inspired basis for developing highly adaptive, distributed computing systems that can, at fine levels of granularity, fault detect, diagnose and self-repair autonomously, without the traditional constraint of a central fault detect/repair unit.
Collapse
Affiliation(s)
- John Wade
- Intelligent Systems Research Center, School of Computing and Intelligent Systems, University of UlsterDerry, Northern Ireland, UK
| | - Liam McDaid
- Intelligent Systems Research Center, School of Computing and Intelligent Systems, University of UlsterDerry, Northern Ireland, UK
| | - Jim Harkin
- Intelligent Systems Research Center, School of Computing and Intelligent Systems, University of UlsterDerry, Northern Ireland, UK
| | - Vincenzo Crunelli
- Neuroscience Division, Cardiff School of Biosciences, University of CardiffCardiff, UK
| | - Scott Kelso
- Intelligent Systems Research Center, School of Computing and Intelligent Systems, University of UlsterDerry, Northern Ireland, UK
- Center for Complex Systems and Brain Sciences, Florida Atlantic UniversityBoca Raton, FL, USA
| |
Collapse
|
33
|
Sebastião AM, Colino-Oliveira M, Assaife-Lopes N, Dias RB, Ribeiro JA. Lipid rafts, synaptic transmission and plasticity: impact in age-related neurodegenerative diseases. Neuropharmacology 2012; 64:97-107. [PMID: 22820274 DOI: 10.1016/j.neuropharm.2012.06.053] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 06/23/2012] [Accepted: 06/26/2012] [Indexed: 10/28/2022]
Abstract
The synapse is a crowded area. In the last years, the concept that proteins can be organized in different membrane domains according to their structure has emerged. Cholesterol-rich membrane domains, or lipid rafts, form an organized portion of the membrane that is thought to concentrate signaling molecules. Accumulating evidence has shown that both the pre-synaptic and post-synaptic sites are highly enriched in lipid rafts, which are likely to organize and maintain synaptic proteins in their precise localization. Here we review recent studies highlighting the importance of lipid rafts for synaptic function and plasticity, as well as their relevance for age or disease-related cognitive impairment. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Ana M Sebastião
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal.
| | | | | | | | | |
Collapse
|
34
|
Abstract
Glioblastoma multiforme (GBM) is the most common malignant brain tumor and is characterized by high invasiveness, poor prognosis, and limited therapeutic options. Biochemical and morphological experiments have shown the presence of caveolae in glioblastoma cells. Caveolae are flask-shaped plasma membrane subdomains that play trafficking, mechanosensing, and signaling roles. Caveolin-1 is a membrane protein that participates in the formation of caveolae and binds a multitude of signaling proteins, compartmentalizing them in caveolae and often directly regulating their activity via binding to its scaffolding domain. Caveolin-1 has been proposed to behave either as a tumor suppressor or as an ongogene depending on the tumor type and progress. This review discusses the existing information on the expression and function of caveolin-1 and caveolae in GBM and the role of this organelle and its defining protein on cellular signaling, growth, and invasiveness of GBM. We further analyze the available data suggesting caveolin-1 could be a target in GBM therapy.
Collapse
Affiliation(s)
- Marie-Odile Parat
- University of Queensland School of Pharmacy, PACE, 20 Cornwall St., Woollloongabba QLD 4102, Australia.
| | | |
Collapse
|
35
|
Takahashi-Iwanaga H, Iwanaga T. Accumulated caveolae constitute subcellular compartments for glial calcium signaling in lanceolate sensory endings innervating rat vibrissae. J Comp Neurol 2012; 520:2053-66. [DOI: 10.1002/cne.23028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Rosenhouse‐Dantsker A, Mehta D, Levitan I. Regulation of Ion Channels by Membrane Lipids. Compr Physiol 2012; 2:31-68. [DOI: 10.1002/cphy.c110001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Wade JJ, McDaid LJ, Harkin J, Crunelli V, Kelso JAS. Bidirectional coupling between astrocytes and neurons mediates learning and dynamic coordination in the brain: a multiple modeling approach. PLoS One 2011; 6:e29445. [PMID: 22242121 PMCID: PMC3248449 DOI: 10.1371/journal.pone.0029445] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 11/28/2011] [Indexed: 11/30/2022] Open
Abstract
In recent years research suggests that astrocyte networks, in addition to nutrient and waste processing functions, regulate both structural and synaptic plasticity. To understand the biological mechanisms that underpin such plasticity requires the development of cell level models that capture the mutual interaction between astrocytes and neurons. This paper presents a detailed model of bidirectional signaling between astrocytes and neurons (the astrocyte-neuron model or AN model) which yields new insights into the computational role of astrocyte-neuronal coupling. From a set of modeling studies we demonstrate two significant findings. Firstly, that spatial signaling via astrocytes can relay a "learning signal" to remote synaptic sites. Results show that slow inward currents cause synchronized postsynaptic activity in remote neurons and subsequently allow Spike-Timing-Dependent Plasticity based learning to occur at the associated synapses. Secondly, that bidirectional communication between neurons and astrocytes underpins dynamic coordination between neuron clusters. Although our composite AN model is presently applied to simplified neural structures and limited to coordination between localized neurons, the principle (which embodies structural, functional and dynamic complexity), and the modeling strategy may be extended to coordination among remote neuron clusters.
Collapse
Affiliation(s)
- John J Wade
- Intelligent Systems Research Centre, School of Computing and Intelligent Systems, University of Ulster, Derry, Northern Ireland.
| | | | | | | | | |
Collapse
|
38
|
Kruglov EA, Gautam S, Guerra MT, Nathanson MH. Type 2 inositol 1,4,5-trisphosphate receptor modulates bile salt export pump activity in rat hepatocytes. Hepatology 2011; 54:1790-9. [PMID: 21748767 PMCID: PMC3205211 DOI: 10.1002/hep.24548] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 06/26/2011] [Indexed: 12/16/2022]
Abstract
UNLABELLED Bile salt secretion is mediated primarily by the bile salt export pump (Bsep), a transporter on the canalicular membrane of the hepatocyte. However, little is known about the short-term regulation of Bsep activity. Ca(2+) regulates targeting and insertion of transporters in many cell systems, and Ca(2+) release near the canalicular membrane is mediated by the type II inositol 1,4,5-trisphosphate receptor (InsP3R2), so we investigated the possible role of InsP3R2 in modulating Bsep activity. The kinetics of Bsep activity were monitored by following secretion of the fluorescent Bsep substrate cholylglycylamido-fluorescein (CGamF) in rat hepatocytes in collagen sandwich culture, an isolated cell system in which structural and functional polarity is preserved. CGamF secretion was nearly eliminated in cells treated with Bsep small interfering RNA (siRNA), demonstrating specificity of this substrate for Bsep. Secretion was also reduced after chelating intracellular calcium, inducing redistribution of InsP3R2 by depleting the cell membrane of cholesterol, or reducing InsP3R function by either knocking down InsP3R2 expression using siRNA or pharmacologic inhibition using xestospongin C. Confocal immunofluorescence showed that InsP3R2 and Bsep are in close proximity in the canalicular region, both in rat liver and in hepatocytes in sandwich culture. However, after knocking down InsP3R2 or inducing its dysfunction with cholesterol depletion, Bsep redistributed intracellularly. Finally, InsP3R2 was lost from the pericanalicular region in animal models of estrogen- and endotoxin-induced cholestasis. CONCLUSION These data provide evidence that pericanalicular calcium signaling mediated by InsP3R2 plays an important role in maintaining bile salt secretion through posttranslational regulation of Bsep, and suggest that loss or redistribution of InsP3R2 may contribute to the pathophysiology of intrahepatic cholestasis.
Collapse
Affiliation(s)
| | | | | | - Michael H. Nathanson
- Address for correspondence: Michael H. Nathanson, Section of Digestive Diseases, Yale University School of Medicine, 333 Cedar Street, TAC S241D, New Haven, CT. 06520-8019, Phone: (203) 785-7312. Fax: (203) 785-7273
| |
Collapse
|
39
|
Abstract
Recently, consciousness research has gained much attention. Indeed, the question at stake is significant: why is the brain not just a computing device, but generates a perception from within? Ambitious endeavors trying to simulate the entire human brain assume that the algorithm will do the trick: as soon as we assemble the brain in a computer and increase the number of operations per time, consciousness will emerge by itself. I disagree with this simplistic representation. My argument emerges from the "atomism paradox": the irreducible space of the consciously perceived world, the endospace is incompatible with the reducible and decomposable architecture of the brain or a computer. I will first discuss the fundamental challenges in current consciousness models and then propose a new model based on the fractality principle: "the whole is in each of its parts". This new model copes with the atomism paradox by implementing an iterative mapping of information from higher order brain structures to smaller scales on the cellular and molecular level, which I will refer to as "fractalization". This information fractalization gives rise to a new form of matter that is conscious ("bright matter"). Bright matter is composed of conscious particles or units named "sentyons". The internal fractality of these sentyons will close a loop (the "psychic loop") in a recurrent fractal neural network (RFNN) that allows for continuous and complete information transformation and sharing between higher order brain structures and the endpoint substrate of consciousness at the molecular level.
Collapse
|
40
|
Paez PM, Fulton D, Spreuer V, Handley V, Campagnoni AT. Modulation of canonical transient receptor potential channel 1 in the proliferation of oligodendrocyte precursor cells by the golli products of the myelin basic protein gene. J Neurosci 2011; 31:3625-37. [PMID: 21389218 PMCID: PMC3076512 DOI: 10.1523/jneurosci.4424-10.2011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 11/22/2010] [Accepted: 01/03/2011] [Indexed: 12/18/2022] Open
Abstract
Golli proteins, products of the myelin basic protein gene, function as a new type of modulator of intracellular Ca(2+) levels in oligodendrocyte progenitor cells (OPCs). Because of this, they affect a number of Ca(2+)-dependent functions, such as OPC migration and process extension. To examine further the Ca(2+) channels regulated by golli, we studied the store-operated Ca(2+) channels (SOCCs) in OPCs and acute brain slice preparations from golli knock-out and golli-overexpressing mice. Our results showed that pharmacologically induced Ca(2+) release from intracellular stores evoked a significant extracellular Ca(2+) entry after store depletion in OPCs. They also indicated that, under these pharmacological conditions, golli promoted activation of Ca(2+) influx by SOCCs in cultured OPCs as well as in tissue slices. The canonical transient receptor potential family of Ca(2+) channels (TRPCs) has been postulated to be SOCC subunits in oligodendrocytes. Using a small interfering RNA knockdown approach, we provided direct evidence that TRPC1 is involved in store-operated Ca(2+) influx in OPCs and that it is modulated by golli. Furthermore, our data indicated that golli is probably associated with TRPC1 at OPC processes. Additionally, we found that TRPC1 expression is essential for the effects of golli on OPC proliferation. In summary, our data indicate a key role for golli proteins in the regulation of TRPC-mediated Ca(2+) influx, a finding that has profound consequences for the regulation of multiple biological processes in OPCs. More important, we have shown that extracellular Ca(2+) uptake through TRPC1 is an essential component in the mechanism of OPC proliferation.
Collapse
Affiliation(s)
- Pablo M Paez
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California at Los Angeles, California 90095-7332, USA.
| | | | | | | | | |
Collapse
|
41
|
Shoeb M, Laloraya M, Kumar PG. Progesterone-induced reorganisation of NOX-2 components in membrane rafts is critical for sperm functioning in Capra hircus. Andrologia 2010; 42:356-65. [DOI: 10.1111/j.1439-0272.2009.01024.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
42
|
Rao W, Isaac RE, Keen JN. An analysis of the Caenorhabditis elegans lipid raft proteome using geLC-MS/MS. J Proteomics 2010; 74:242-53. [PMID: 21070894 DOI: 10.1016/j.jprot.2010.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 10/20/2010] [Accepted: 11/02/2010] [Indexed: 11/16/2022]
Abstract
Lipid rafts are microdomains of the phospholipid bilayer, proposed to form semi-stable "islands" that act as a platform for several important cellular processes; major classes of raft-resident proteins include signalling proteins and glycosylphosphatidylinositol (GPI)-anchored proteins. Proteomic studies into lipid rafts have been mainly carried out in mammalian cell lines and single cell organisms. The nematode Caenorhabditis elegans, the model organism with a well-defined developmental profile, is ideally suited for the study of this subcellular locale in a complex developmental context. A study of the lipid raft proteome of C. elegans is presented here. A total of 44 proteins were identified from the lipid raft fraction using geLC-MS/MS, of which 40 have been determined to be likely raft proteins after analysis of predicted functions. Prediction of GPI-anchoring of the proteins found 21 to be potentially modified in this way, two of which were experimentally confirmed to be GPI-anchored. This work is the first reported study of the lipid raft proteome in C. elegans. The results show that raft proteins, including numerous GPI-anchored proteins, may have a variety of potentially important roles within the nematode, and will hopefully lead to C. elegans becoming a useful model for the study of lipid rafts.
Collapse
Affiliation(s)
- Wei Rao
- Institute of Integrative and Comparative Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | | |
Collapse
|
43
|
Morita M, Kudo Y. Growth factors upregulate astrocyte [Ca2+]i oscillation by increasing SERCA2b expression. Glia 2010; 58:1988-95. [DOI: 10.1002/glia.21067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
44
|
Schmitz M, Klöppner S, Klopfleisch S, Möbius W, Schwartz P, Zerr I, Althaus HH. Mutual effects of caveolin and nerve growth factor signaling in pig oligodendrocytes. J Neurosci Res 2010; 88:572-88. [PMID: 19795378 DOI: 10.1002/jnr.22235] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signaling of growth factors may depend on the recruitment of their receptors to specialized microdomains. Previous reports on PC12 cells indicated an interaction of raft-organized caveolin and TrkA signaling. Because porcine oligodendrocytes (OLs) respond to nerve growth factor (NGF), we were interested to know whether caveolin also plays a role in oligodendroglial NGF/TrkA signaling. OLs expressed caveolin at the plasma membrane but also intracellularly. This was partially organized in the classically Omega-shaped invaginations, which may represent caveolae. We could show that caveolin and TrkA colocalize by using a discontinuous sucrose gradient (Song et al. [1996] J. Biol. Chem. 271:9690-9697), MACS technology, and immunoprecipitation. However, differential extraction of caveolin and TrkA with Triton X-100 at 4 degrees C indicated that caveolin and TrkA are probably not exclusively present in detergent-resistant, caveolin-containing rafts (CCRs). NGF treatment of OLs up-regulated the expression of caveolin-1 (cav-1) and stimulated tyrosine-14 phosphorylation of cav-1. Furthermore, OLs were transfected with cav-1-specific small interfering RNA (siRNA). A knockdown of cav-1 resulted in a reduced activation of downstream components of the NGF signaling cascade, such as p21Ras and mitogen-activated protein kinase (MAPK) after NGF exposure of OLs. Subsequently, increased oligodendroglial process formation via NGF was impaired. The present study indicates that CCRs/caveolin could play a modulating role during oligodendroglial differentiation and regeneration.
Collapse
Affiliation(s)
- Matthias Schmitz
- RU Neural Regeneration, Max-Planck Institute of Experimental Medicine, Goettingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
45
|
Pessah IN, Cherednichenko G, Lein PJ. Minding the calcium store: Ryanodine receptor activation as a convergent mechanism of PCB toxicity. Pharmacol Ther 2010; 125:260-85. [PMID: 19931307 PMCID: PMC2823855 DOI: 10.1016/j.pharmthera.2009.10.009] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 10/30/2009] [Indexed: 11/24/2022]
Abstract
Chronic low-level polychlorinated biphenyl (PCB) exposures remain a significant public health concern since results from epidemiological studies indicate that PCB burden is associated with immune system dysfunction, cardiovascular disease, and impairment of the developing nervous system. Of these various adverse health effects, developmental neurotoxicity has emerged as a particularly vulnerable endpoint in PCB toxicity. Arguably the most pervasive biological effects of PCBs could be mediated by their ability to alter the spatial and temporal fidelity of Ca2+ signals through one or more receptor-mediated processes. This review will focus on our current knowledge of the structure and function of ryanodine receptors (RyRs) in muscle and nerve cells and how PCBs and related non-coplanar structures alter these functions. The molecular and cellular mechanisms by which non-coplanar PCBs and related structures alter local and global Ca2+ signaling properties and the possible short and long-term consequences of these perturbations on neurodevelopment and neurodegeneration are reviewed.
Collapse
Affiliation(s)
- Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | | | | |
Collapse
|
46
|
Martín C, Gómez-Bilbao G, Ostolaza H. Bordetella adenylate cyclase toxin promotes calcium entry into both CD11b+ and CD11b- cells through cAMP-dependent L-type-like calcium channels. J Biol Chem 2009; 285:357-64. [PMID: 19875442 DOI: 10.1074/jbc.m109.003491] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adenylate cyclase toxin (ACT), a 200 kDa protein, is an essential virulence factor for Bordetella pertussis, the bacterium that causes whooping cough. ACT is a member of the pore-forming RTX (repeats-in-toxin) family of proteins that share a characteristic calcium-binding motif of Gly- and Asp-rich nonapeptide repeats and a marked cytolytic or cytotoxic activity. In addition, ACT exhibits a distinctive feature: it has an N-terminal calmodulin-dependent adenylate cyclase domain. Translocation of this domain into the host cytoplasm results in uncontrolled production of cAMP, and it has classically been assumed that this surge in cAMP is the basis for the toxin-mediated killing. Several members of the RTX family of toxins, including ACT, have been shown to induce intracellular calcium increases, through different mechanisms. We show here that ACT stimulates a raft-mediated calcium influx, through its cAMP production activity, that activates PKA, which in turn activates calcium channels with L-type properties. This process is shown to occur both in CD11b(+) and CD11b(-) cells, suggesting a common mechanism, independent of the toxin receptor. We also show that this ACT-induced calcium influx does not correlate with the toxin-induced cytotoxicity.
Collapse
Affiliation(s)
- César Martín
- Unidad de Biofísica, Departamento de Bioquímica, Universidad del País Vasco, Centro Mixto CSIC-UPV/EHU, 48080 Bilbao, Spain
| | | | | |
Collapse
|
47
|
Abstract
Rafts are domains of the plasma membrane, enriched in cholesterol and sphingolipids; they form a platform for signaling proteins and receptors. The lipid rafts are utilized in the replication cycle of numerous viruses. Internalization receptors of many viruses localize to rafts or are recruited there after virus binding. Arrays of signal transduction proteins found in rafts contribute to efficient trafficking and productive infection. Some viruses are dependent on raft domains for the biogenesis of their membranous replication structures. Finally, rafts are often important in virus assembly and budding. Subsequently, raft components in the viral envelope may be vital for the entry to a new host cell. Here, we summarize the current knowledge of the involvement of rafts in virus infection.
Collapse
Affiliation(s)
- Paula Upla
- Department of Biological & Environmental Science/Nanoscience Center, University of Jyväskylä, FI-40351 Jyväskylä, Finland
| | - Timo Hyypiä
- Department of Virology, University of Turku, FI-20520 Turku, Finland
| | - Varpu Marjomäki
- Department of Biological & Environmental Science/Nanoscience Center, University of Jyväskylä, FI-40351 Jyväskylä, Finland
| |
Collapse
|
48
|
Wojcikiewicz RJH, Pearce MMP, Sliter DA, Wang Y. When worlds collide: IP(3) receptors and the ERAD pathway. Cell Calcium 2009; 46:147-53. [PMID: 19709743 DOI: 10.1016/j.ceca.2009.05.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 05/01/2009] [Accepted: 05/05/2009] [Indexed: 12/13/2022]
Abstract
While cell signaling devotees tend to think of the endoplasmic reticulum (ER) as a Ca(2+) store, those who study protein synthesis tend to see it more as site for protein maturation, or even degradation when proteins do not fold properly. These two worldviews collide when inositol 1,4,5-trisphosphate (IP(3)) receptors are activated, since in addition to acting as release channels for stored ER Ca(2+), IP(3) receptors are rapidly destroyed via the ER-associated degradation (ERAD) pathway, a ubiquitination- and proteasome-dependent mechanism that clears the ER of aberrant proteins. Here we review recent studies showing that activated IP(3) receptors are ubiquitinated in an unexpectedly complex manner, and that a novel complex composed of the ER membrane proteins SPFH1 and SPFH2 (erlin 1 and 2) binds to IP(3) receptors immediately after they are activated and mediates their ERAD. Remarkably, it seems that the conformational changes that underpin channel opening make IP(3) receptors resemble aberrant proteins, which triggers their binding to the SPFH1/2 complex, their ubiquitination and extraction from the ER membrane and finally, their degradation by the proteasome. This degradation of activated IP(3) receptors by the ERAD pathway serves to reduce the sensitivity of ER Ca(2+) stores to IP(3) and may protect cells against deleterious effects of over-activation of Ca(2+) signaling pathways.
Collapse
|
49
|
FcgammaRI ligation leads to a complex with BLT1 in lipid rafts that enhances rat lung macrophage antimicrobial functions. Blood 2009; 114:3316-24. [PMID: 19657115 PMCID: PMC2759654 DOI: 10.1182/blood-2009-01-199919] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leukotriene (LT) B(4) is generated in response to engagement of the Fc gamma receptor (Fc gamma R) and potently contributes to Fc gamma R-mediated antimicrobial functions in pulmonary alveolar macrophages. In this study, we report that the LTB(4) receptor leukotriene B(4) receptor 1 (BLT1) redistributes from nonlipid raft (LR) to LR membrane microdomains upon immunoglobulin G-red blood cell, but not LTB(4), challenge. Cholesterol depletion to disrupt LRs abolished LTB(4)-induced enhancement of phagocytosis, microbicidal activity, and signaling. The dependence on LR integrity for BLT1 signaling correlated with formation of a complex consisting of BLT1, its primary coupled G protein G alpha i3, Src kinase, and Fc gamma RI within LRs. This association was dependent on Src-mediated phosphorylation of BLT1. These data identify a novel form of regulation in which engagement of a macrophage immunoreceptor recruits a stimulatory G protein-coupled receptor into a LR microdomain with resultant enhanced antimicrobial signaling.
Collapse
|
50
|
Monastyrskaya K, Babiychuk EB, Draeger A. The annexins: spatial and temporal coordination of signaling events during cellular stress. Cell Mol Life Sci 2009; 66:2623-42. [PMID: 19381436 PMCID: PMC11115530 DOI: 10.1007/s00018-009-0027-1] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Revised: 02/09/2009] [Accepted: 03/27/2009] [Indexed: 12/15/2022]
Abstract
Annexins are a family of structurally related, Ca2+-sensitive proteins that bind to negatively charged phospholipids and establish specific interactions with other lipids and lipid microdomains. They are present in all eukaryotic cells and share a common folding motif, the "annexin core", which incorporates Ca2+- and membrane-binding sites. Annexins participate in a variety of intracellular processes, ranging from the regulation of membrane dynamics to cell migration, proliferation, and apoptosis. Here we focus on the role of annexins in cellular signaling during stress. A chronic stress response triggers the activation of different intracellular pathways, resulting in profound changes in Ca2+ and pH homeostasis and the production of lipid second messengers. We review the latest data on how these changes are sensed by the annexins, which have the ability to simultaneously interact with specific lipid and protein moieties at the plasma membrane, contributing to stress adaptation via regulation of various signaling pathways.
Collapse
Affiliation(s)
- Katia Monastyrskaya
- Department of Cell Biology, Institute of Anatomy, University of Bern, 3000 Bern 9, Switzerland.
| | | | | |
Collapse
|