1
|
Stiekema M, Houben F, Verheyen F, Borgers M, Menzel J, Meschkat M, van Zandvoort MAMJ, Ramaekers FCS, Broers JLV. The Role of Lamins in the Nucleoplasmic Reticulum, a Pleiomorphic Organelle That Enhances Nucleo-Cytoplasmic Interplay. Front Cell Dev Biol 2022; 10:914286. [PMID: 35784476 PMCID: PMC9243388 DOI: 10.3389/fcell.2022.914286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/24/2022] [Indexed: 12/15/2022] Open
Abstract
Invaginations of the nuclear membrane occur in different shapes, sizes, and compositions. Part of these pleiomorphic invaginations make up the nucleoplasmic reticulum (NR), while others are merely nuclear folds. We define the NR as tubular invaginations consisting of either both the inner and outer nuclear membrane, or only the inner nuclear membrane. Specifically, invaginations of both the inner and outer nuclear membrane are also called type II NR, while those of only the inner nuclear membrane are defined as type I NR. The formation and structure of the NR is determined by proteins associated to the nuclear membrane, which induce a high membrane curvature leading to tubular invaginations. Here we review and discuss the current knowledge of nuclear invaginations and the NR in particular. An increase in tubular invaginations of the nuclear envelope is associated with several pathologies, such as laminopathies, cancer, (reversible) heart failure, and Alzheimer’s disease. Furthermore, viruses can induce both type I and II NR. In laminopathies, the amount of A-type lamins throughout the nucleus is generally decreased or the organization of lamins or lamin-associated proteins is disturbed. Also, lamin overexpression or modulation of lamin farnesylation status impacts NR formation, confirming the importance of lamin processing in NR formation. Virus infections reorganize the nuclear lamina via (de)phosphorylation of lamins, leading to an uneven thickness of the nuclear lamina and in turn lobulation of the nuclear membrane and the formation of invaginations of the inner nuclear membrane. Since most studies on the NR have been performed with cell cultures, we present additional proof for the existence of these structures in vivo, focusing on a variety of differentiated cardiovascular and hematopoietic cells. Furthermore, we substantiate the knowledge of the lamin composition of the NR by super-resolution images of the lamin A/C and B1 organization. Finally, we further highlight the essential role of lamins in NR formation by demonstrating that (over)expression of lamins can induce aberrant NR structures.
Collapse
Affiliation(s)
- Merel Stiekema
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Frederik Houben
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Healthcare, PXL University College, Hasselt, Belgium
| | - Fons Verheyen
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Marcel Borgers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | | | - Marc A. M. J. van Zandvoort
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research IMCAR, RWTH Aachen University, Aachen, Germany
| | - Frans C. S. Ramaekers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jos L. V. Broers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands
- *Correspondence: Jos L. V. Broers,
| |
Collapse
|
2
|
Corbeil D, Santos MF, Karbanová J, Kurth T, Rappa G, Lorico A. Uptake and Fate of Extracellular Membrane Vesicles: Nucleoplasmic Reticulum-Associated Late Endosomes as a New Gate to Intercellular Communication. Cells 2020; 9:cells9091931. [PMID: 32825578 PMCID: PMC7563309 DOI: 10.3390/cells9091931] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular membrane vesicles (EVs) are emerging as new vehicles in intercellular communication, but how the biological information contained in EVs is shared between cells remains elusive. Several mechanisms have been described to explain their release from donor cells and the initial step of their uptake by recipient cells, which triggers a cellular response. Yet, the intracellular routes and subcellular fate of EV content upon internalization remain poorly characterized. This is particularly true for EV-associated proteins and nucleic acids that shuttle to the nucleus of host cells. In this review, we will describe and discuss the release of EVs from donor cells, their uptake by recipient cells, and the fate of their cargoes, focusing on a novel intracellular route wherein small GTPase Rab7+ late endosomes containing endocytosed EVs enter into nuclear envelope invaginations and deliver their cargo components to the nucleoplasm of recipient cells. A tripartite protein complex composed of (VAMP)-associated protein A (VAP-A), oxysterol-binding protein (OSBP)-related protein-3 (ORP3), and Rab7 is essential for the transfer of EV-derived components to the nuclear compartment by orchestrating the particular localization of late endosomes in the nucleoplasmic reticulum.
Collapse
Affiliation(s)
- Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-49, 01307 Dresden, Germany; (J.K.)
- Correspondence: (D.C.); (A.L.); Tel.: +49-(0)351-463-40118 (D.C.); +1-(702)-777-3942 (A.L.); Fax: +49-(0)351-463-40244 (D.C.); +1-(702)-777-1758 (A.L.)
| | - Mark F. Santos
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
| | - Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47-49, 01307 Dresden, Germany; (J.K.)
| | - Thomas Kurth
- Center for Regenerative Therapies Dresden and CMCB, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; (T.K.)
| | - Germana Rappa
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
| | - Aurelio Lorico
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV 89014, USA; (M.F.S.); (G.R.)
- Mediterranean Institute of Oncology, Via Penninazzo, 11, 95029 Viagrande, Italy
- Correspondence: (D.C.); (A.L.); Tel.: +49-(0)351-463-40118 (D.C.); +1-(702)-777-3942 (A.L.); Fax: +49-(0)351-463-40244 (D.C.); +1-(702)-777-1758 (A.L.)
| |
Collapse
|
3
|
Dewenter M, von der Lieth A, Katus HA, Backs J. Calcium Signaling and Transcriptional Regulation in Cardiomyocytes. Circ Res 2017; 121:1000-1020. [DOI: 10.1161/circresaha.117.310355] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Calcium (Ca
2+
) is a universal regulator of various cellular functions. In cardiomyocytes, Ca
2+
is the central element of excitation–contraction coupling, but also impacts diverse signaling cascades and influences the regulation of gene expression, referred to as excitation–transcription coupling. Disturbances in cellular Ca
2+
-handling and alterations in Ca
2+
-dependent gene expression patterns are pivotal characteristics of failing cardiomyocytes, with several excitation–transcription coupling pathways shown to be critically involved in structural and functional remodeling processes. Thus, targeting Ca
2+
-dependent transcriptional pathways might offer broad therapeutic potential. In this article, we (1) review cytosolic and nuclear Ca
2+
dynamics in cardiomyocytes with respect to their impact on Ca
2+
-dependent signaling, (2) give an overview on Ca
2+
-dependent transcriptional pathways in cardiomyocytes, and (3) discuss implications of excitation–transcription coupling in the diseased heart.
Collapse
Affiliation(s)
- Matthias Dewenter
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Albert von der Lieth
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Hugo A. Katus
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Johannes Backs
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| |
Collapse
|
4
|
Torgan CE, Daniels MP. Calcineurin Localization in Skeletal Muscle Offers Insights into Potential New Targets. J Histochem Cytochem 2016; 54:119-28. [PMID: 16174789 DOI: 10.1369/jhc.5a6769.2005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The Ca2+/calmodulin-activated protein phosphatase, calcineurin, is believed to regulate the development and function of skeletal and cardiac muscle. Striated muscle contains many calcineurin substrates, a few of which have been colocalized or found in molecular complexes with calcineurin. We examined the subcellular distribution of calcineurin in developing rat skeletal muscle cells and adult mouse skeletal muscle fibers by immunofluorescence microscopy. We found low levels of calcineurin immunoreactivity in the cytoplasm of myoblasts and higher levels in cytoplasmic vesicles of myotubes. Most of these vesicles were not immunoreactive for ryanodine receptors and, those that were, represented a small fraction of nascent triad junctions. In adult myofibers, calcineurin was largely associated with triads. Weaker calcineurin immunoreactivity occurred in the sarcoplasmic reticulum at the level of the M line. Unexpectedly, we found tiny clusters of calcineurin associated with nucleoli of developing myofiber nuclei. There were one to three clusters per nucleolus, either within or at the edges of fibrillar centers where ribosomal genes are transcribed. This suggests a role for calcineurin in regulating ribosome synthesis. Our findings suggest a variety of potential new targets and pathways through which calcineurin could regulate skeletal muscle development and plasticity and underscore the importance of spatial specificity in this regulation.
Collapse
Affiliation(s)
- Carol E Torgan
- Laboratory of Cell Biology, National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, Bethesda, Maryland 20892-8017, USA
| | | |
Collapse
|
5
|
McEntagart M, Williamson KA, Rainger JK, Wheeler A, Seawright A, De Baere E, Verdin H, Bergendahl LT, Quigley A, Rainger J, Dixit A, Sarkar A, López Laso E, Sanchez-Carpintero R, Barrio J, Bitoun P, Prescott T, Riise R, McKee S, Cook J, McKie L, Ceulemans B, Meire F, Temple IK, Prieur F, Williams J, Clouston P, Németh AH, Banka S, Bengani H, Handley M, Freyer E, Ross A, van Heyningen V, Marsh JA, Elmslie F, FitzPatrick DR. A Restricted Repertoire of De Novo Mutations in ITPR1 Cause Gillespie Syndrome with Evidence for Dominant-Negative Effect. Am J Hum Genet 2016; 98:981-992. [PMID: 27108798 PMCID: PMC4863663 DOI: 10.1016/j.ajhg.2016.03.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/16/2016] [Indexed: 12/19/2022] Open
Abstract
Gillespie syndrome (GS) is characterized by bilateral iris hypoplasia, congenital hypotonia, non-progressive ataxia, and progressive cerebellar atrophy. Trio-based exome sequencing identified de novo mutations in ITPR1 in three unrelated individuals with GS recruited to the Deciphering Developmental Disorders study. Whole-exome or targeted sequence analysis identified plausible disease-causing ITPR1 mutations in 10/10 additional GS-affected individuals. These ultra-rare protein-altering variants affected only three residues in ITPR1: Glu2094 missense (one de novo, one co-segregating), Gly2539 missense (five de novo, one inheritance uncertain), and Lys2596 in-frame deletion (four de novo). No clinical or radiological differences were evident between individuals with different mutations. ITPR1 encodes an inositol 1,4,5-triphosphate-responsive calcium channel. The homo-tetrameric structure has been solved by cryoelectron microscopy. Using estimations of the degree of structural change induced by known recessive- and dominant-negative mutations in other disease-associated multimeric channels, we developed a generalizable computational approach to indicate the likely mutational mechanism. This analysis supports a dominant-negative mechanism for GS variants in ITPR1. In GS-derived lymphoblastoid cell lines (LCLs), the proportion of ITPR1-positive cells using immunofluorescence was significantly higher in mutant than control LCLs, consistent with an abnormality of nuclear calcium signaling feedback control. Super-resolution imaging supports the existence of an ITPR1-lined nucleoplasmic reticulum. Mice with Itpr1 heterozygous null mutations showed no major iris defects. Purkinje cells of the cerebellum appear to be the most sensitive to impaired ITPR1 function in humans. Iris hypoplasia is likely to result from either complete loss of ITPR1 activity or structure-specific disruption of multimeric interactions.
Collapse
Affiliation(s)
- Meriel McEntagart
- Medical Genetics, St George's University Hospitals NHS Foundation Trust, Cranmer Terrace, London SW17 0RE, UK
| | - Kathleen A Williamson
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Jacqueline K Rainger
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Ann Wheeler
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Anne Seawright
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Elfride De Baere
- Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Medical Research Building (MRB), 1st Floor, Room 110.029, De Pintelaan 185, 9000 Ghent, Belgium
| | - Hannah Verdin
- Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Medical Research Building (MRB), 1st Floor, Room 110.029, De Pintelaan 185, 9000 Ghent, Belgium
| | - L Therese Bergendahl
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Alan Quigley
- Department of Radiology, Royal Hospital for Sick Children, Edinburgh EH9 1LF, UK
| | - Joe Rainger
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK; Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Abhijit Dixit
- Clinical Genetics, Nottingham City Hospital, Hucknall Road, Nottingham NG5 1PB, UK
| | - Ajoy Sarkar
- Clinical Genetics, Nottingham City Hospital, Hucknall Road, Nottingham NG5 1PB, UK
| | - Eduardo López Laso
- Pediatric Neurology Unit, Department of Pediatrics, Reina Sofia University Hospital, Av. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | - Rocio Sanchez-Carpintero
- Paediatric Neurology Unit, Department of Paediatrics, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jesus Barrio
- Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Pierre Bitoun
- Service de pédiatrie, CHU Paris Seine-Saint-Denis - Hôpital Jean Verdier Avenue du 14 juillet, 93140 Bondy, France
| | - Trine Prescott
- Department of Medical Genetics, Oslo University Hospital, 0424 Oslo, Norway
| | - Ruth Riise
- Department of Ophthalmology, Innland Hospital, 2418 Elverum, Norway
| | - Shane McKee
- Northern Ireland Regional Genetics Service, Belfast City Hospital, Belfast BT9 7AB, UK
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Western Bank, Sheffield S10 2TH, UK
| | - Lisa McKie
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Berten Ceulemans
- Department of Neurology-Pediatric Neurology, University and University Hospital Antwerp, Antwerp 2650, Belgium
| | - Françoise Meire
- Department of Ophthalmology, Queen Fabiola Children's University Hospital, 1020 Brussels, Belgium
| | - I Karen Temple
- Human Development and Health Academic Unit, University Hospital Southampton, Tremona Road, University of Southampton, Southampton SO16 6YD, UK
| | - Fabienne Prieur
- Service Génétique, Plateau de biologie, CHU Saint Etienne, 42055 Saint Etienne cedex 2, France
| | - Jonathan Williams
- Oxford University Hospitals NHS Trust, Oxford Medical Genetics Laboratories, The Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - Penny Clouston
- Oxford University Hospitals NHS Trust, Oxford Medical Genetics Laboratories, The Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - Andrea H Németh
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 7LJ, UK
| | - Siddharth Banka
- Manchester Centre for Genomic Medicine, University of Manchester, St. Mary's Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Hemant Bengani
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Mark Handley
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Elisabeth Freyer
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Allyson Ross
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Veronica van Heyningen
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Joseph A Marsh
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Frances Elmslie
- Medical Genetics, St George's University Hospitals NHS Foundation Trust, Cranmer Terrace, London SW17 0RE, UK
| | - David R FitzPatrick
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK.
| |
Collapse
|
6
|
Ibarra C, Vicencio JM, Varas-Godoy M, Jaimovich E, Rothermel BA, Uhlén P, Hill JA, Lavandero S. An integrated mechanism of cardiomyocyte nuclear Ca(2+) signaling. J Mol Cell Cardiol 2014; 75:40-8. [PMID: 24997440 PMCID: PMC4626248 DOI: 10.1016/j.yjmcc.2014.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 06/11/2014] [Accepted: 06/26/2014] [Indexed: 01/05/2023]
Abstract
In cardiomyocytes, Ca(2+) plays a central role in governing both contraction and signaling events that regulate gene expression. Current evidence indicates that discrimination between these two critical functions is achieved by segregating Ca(2+) within subcellular microdomains: transcription is regulated by Ca(2+) release within nuclear microdomains, and excitation-contraction coupling is regulated by cytosolic Ca(2+). Accordingly, a variety of agonists that control cardiomyocyte gene expression, such as endothelin-1, angiotensin-II or insulin-like growth factor-1, share the feature of triggering nuclear Ca(2+) signals. However, signaling pathways coupling surface receptor activation to nuclear Ca(2+) release, and the phenotypic responses to such signals, differ between agonists. According to earlier hypotheses, the selective control of nuclear Ca(2+) signals by activation of plasma membrane receptors relies on the strategic localization of inositol trisphosphate receptors at the nuclear envelope. There, they mediate Ca(2+) release from perinuclear Ca(2+) stores upon binding of inositol trisphosphate generated in the cytosol, which diffuses into the nucleus. More recently, identification of such receptors at nuclear membranes or perinuclear sarcolemmal invaginations has uncovered novel mechanisms whereby agonists control nuclear Ca(2+) release. In this review, we discuss mechanisms for the selective control of nuclear Ca(2+) signals with special focus on emerging models of agonist receptor activation.
Collapse
Affiliation(s)
- Cristián Ibarra
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development, AstraZeneca R&D, Mölndal, Sweden.
| | - Jose Miguel Vicencio
- Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Manuel Varas-Godoy
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Enrique Jaimovich
- Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Joseph A Hill
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sergio Lavandero
- Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
7
|
Baranov MS, Solntsev KM, Baleeva NS, Mishin AS, Lukyanov SA, Lukyanov KA, Yampolsky IV. Red-shifted fluorescent aminated derivatives of a conformationally locked GFP chromophore. Chemistry 2014; 20:13234-41. [PMID: 25171432 DOI: 10.1002/chem.201403678] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Indexed: 11/09/2022]
Abstract
A novel class of fluorescent dyes based on conformationally locked GFP chromophore is reported. These dyes are characterized by red-shifted spectra, high fluorescence quantum yields and pH-independence in physiological pH range. The intra- and intermolecular mechanisms of radiationless deactivation of ABDI-BF2 fluorophore by selective structural locking of various conformational degrees of freedom were studied. A unique combination of solvatochromic and lipophilic properties together with "infinite" photostability (due to a dynamic exchange between free and bound dye) makes some of the novel dyes promising bioinspired tools for labeling cellular membranes, lipid drops and other organelles.
Collapse
Affiliation(s)
- Mikhail S Baranov
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow (Russia).
| | | | | | | | | | | | | |
Collapse
|
8
|
Korfali N, Wilkie GS, Swanson SK, Srsen V, de Las Heras J, Batrakou DG, Malik P, Zuleger N, Kerr ARW, Florens L, Schirmer EC. The nuclear envelope proteome differs notably between tissues. Nucleus 2012; 3:552-64. [PMID: 22990521 PMCID: PMC3515538 DOI: 10.4161/nucl.22257] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
One hypothesis to explain how mutations in the same nuclear envelope proteins yield pathologies focused in distinct tissues is that as yet unidentified tissue-specific partners mediate the disease pathologies. The nuclear envelope proteome was recently determined from leukocytes and muscle. Here the same methodology is applied to liver and a direct comparison of the liver, muscle and leukocyte data sets is presented. At least 74 novel transmembrane proteins identified in these studies have been directly confirmed at the nuclear envelope. Within this set, RT-PCR, western blot and staining of tissue cryosections confirms that the protein complement of the nuclear envelope is clearly distinct from one tissue to another. Bioinformatics reveals similar divergence between tissues across the larger data sets. For proteins acting in complexes according to interactome data, the whole complex often exhibited the same tissue-specificity. Other tissue-specific nuclear envelope proteins identified were known proteins with functions in signaling and gene regulation. The high tissue specificity in the nuclear envelope likely underlies the complex disease pathologies and argues that all organelle proteomes warrant re-examination in multiple tissues.
Collapse
Affiliation(s)
- Nadia Korfali
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Mauger JP. Role of the nuclear envelope in calcium signalling. Biol Cell 2011; 104:70-83. [PMID: 22188206 DOI: 10.1111/boc.201100103] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 11/18/2011] [Indexed: 12/21/2022]
Abstract
The endoplasmic reticulum (ER) is the major Ca(2+) store inside the cell. Its organisation in specialised subdomains allows the local delivery of Ca(2+) to specific cell areas on stimulation. The nuclear envelope (NE), which is continuous with the ER, has a double role: it insulates the nucleoplasm from the cytoplasm and it stores Ca(2+) around the nucleus. Furthermore, all the constituents of the signalling cascade leading to Ca(2+) mobilisation are found in the NE; this allows the nuclear Ca(2+) to be regulated autonomously. On the other hand, cytosolic Ca(2+) transients can propagate within the nucleus via the nuclear pore complex. The variations in nuclear Ca(2+) concentration are important for controlling gene transcription and progression in the cell cycle. Recent data suggest that invaginations of the NE modify the morphology of the nucleus and may affect Ca(2+) dynamics in the nucleus and regulate transcriptional activity.
Collapse
|
10
|
Nalaskowski MM, Fliegert R, Ernst O, Brehm MA, Fanick W, Windhorst S, Lin H, Giehler S, Hein J, Lin YN, Mayr GW. Human inositol 1,4,5-trisphosphate 3-kinase isoform B (IP3KB) is a nucleocytoplasmic shuttling protein specifically enriched at cortical actin filaments and at invaginations of the nuclear envelope. J Biol Chem 2011; 286:4500-10. [PMID: 21148483 PMCID: PMC3039344 DOI: 10.1074/jbc.m110.173062] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 12/02/2010] [Indexed: 11/06/2022] Open
Abstract
Recent studies have shown that inositol 1,4,5-trisphosphate 3-kinase isoform B (IP3KB) possesses important roles in the development of immune cells. IP3KB can be targeted to multiple cellular compartments, among them nuclear localization and binding in close proximity to the plasma membrane. The B isoform is the only IP3K that is almost ubiquitously expressed in mammalian cells. Detailed mechanisms of its targeting regulation will be important in understanding the role of Ins(1,4,5)P(3) phosphorylation on subcellular calcium signaling and compartment-specific initiation of pathways leading to regulatory active higher phosphorylated inositol phosphates. Here, we identified an exportin 1-dependent nuclear export signal ((134)LQRELQNVQV) and characterized the amino acids responsible for nuclear localization of IP3KB ((129)RKLR). These two targeting domains regulate the amount of nuclear IP3KB in cells. We also demonstrated that the localization of IP3KB at the plasma membrane is due to its binding to cortical actin structures. Intriguingly, all three of these targeting activities reside in one small polypeptide segment (amino acids 104-165), which acts as a multitargeting domain (MTD). Finally, a hitherto unknown subnuclear localization of IP3KB could be demonstrated in rapidly growing H1299 cells. IP3KB is specifically enriched at nuclear invaginations extending perpendicular between the apical and basal surface of the nucleus of these flat cells. Such nuclear invaginations are known to be involved in Ins(1,4,5)P(3)-mediated Ca(2+) signaling of the nucleus. Our findings indicate that IP3KB not only regulates cytoplasmic Ca(2+) signals by phosphorylation of subplasmalemmal and cytoplasmic Ins(1,4,5)P(3) but may also be involved in modulating nuclear Ca(2+) signals generated from these nuclear envelope invaginations.
Collapse
Affiliation(s)
- Marcus M Nalaskowski
- Institute of Biochemistry and Molecular Biology I-Cellular Signal Transduction, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Olins AL, Langhans M, Monestier M, Schlotterer A, Robinson DG, Viotti C, Zentgraf H, Zwerger M, Olins DE. An epichromatin epitope: persistence in the cell cycle and conservation in evolution. Nucleus 2011; 2:47-60. [PMID: 21647299 PMCID: PMC3104809 DOI: 10.4161/nucl.2.1.13271] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 09/15/2010] [Accepted: 09/16/2010] [Indexed: 11/19/2022] Open
Abstract
Interphase nuclear architecture is disrupted and rapidly reformed with each cell division cycle. Successive cell generations exhibit a "memory" of this nuclear architecture, as well as for gene expression. Furthermore, many features of nuclear and mitotic chromosome structure are recognizably species and tissue specific. We wish to know what properties of the underlying chromatin structure may determine these conserved features of nuclear architecture. Employing a particular mouse autoimmune anti-nucleosome monoclonal antibody (PL2-6), combined with deconvolution immunofluorescence microscopy, we present evidence for a unique epitope (involving a ternary complex of histones H2A and H2B and DNA) which is localized only at the exterior chromatin surface of interphase nuclei and mitotic chromosomes in mammalian, invertebrate and plant systems. As only the surface chromatin region is identified with antibody PL2-6, we have assigned it the name "epichromatin". We describe an "epichromatin hypothesis", suggesting that epichromatin may have a unique evolutionary conserved conformation which facilitates interaction with the reforming post-mitotic nuclear envelope and a rapid return of interphase nuclear architecture.
Collapse
Affiliation(s)
- Ada L Olins
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME USA. ted proteins (ARPs), a
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Escobar M, Cardenas C, Colavita K, Petrenko NB, Franzini-Armstrong C. Structural evidence for perinuclear calcium microdomains in cardiac myocytes. J Mol Cell Cardiol 2010; 50:451-9. [PMID: 21147122 DOI: 10.1016/j.yjmcc.2010.11.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 11/24/2010] [Accepted: 11/29/2010] [Indexed: 10/18/2022]
Abstract
At each heartbeat, cardiac myocytes are activated by a cytoplasmic Ca(2+) transient in great part due to Ca(2+) release from the sarcoplasmic reticulum via ryanodine receptors (RyRs) clustered within calcium release units (peripheral couplings/dyads). A Ca(2+) transient also occurs in the nucleoplasm, following the cytoplasmic transient with some delay. Under conditions where the InsP3 production is stimulated, these Ca(2+) transients are regulated actively, presumably by an additional release of Ca(2+) via InsP3 receptors (InsP3Rs). This raises the question whether InsP3Rs are appropriately located for this effect and whether sources of InsP3 and Ca(2+) are available for their activation. We have defined the structural basis for InsP3R activity at the nucleus, using immunolabeling for confocal microscopy and freeze-drying/shadowing, T tubule "staining" and thin sectioning for electron microscopy. By these means we establish the presence of InsP3R at the outer nuclear envelope and show a close spatial relationship between the nuclear envelope, T tubules (a likely source of InsP3) and dyads (the known source of Ca(2+)). The frequency, distribution and distance from the nucleus of T tubules and dyads appropriately establish local perinuclear Ca(2+) microdomains in cardiac myocytes.
Collapse
Affiliation(s)
- Matias Escobar
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | | | | | | | | |
Collapse
|
13
|
Alonso MT, García-Sancho J. Nuclear Ca(2+) signalling. Cell Calcium 2010; 49:280-9. [PMID: 21146212 DOI: 10.1016/j.ceca.2010.11.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 10/30/2010] [Accepted: 11/08/2010] [Indexed: 10/18/2022]
Abstract
Ca(2+) signalling is important for controlling gene transcription. Changes of the cytosolic Ca(2+) ([Ca(2+)](C)) may promote migration of transcription factors or transcriptional regulators to the nucleus. Changes of the nucleoplasmic Ca(2+) ([Ca(2+)](N)) can also regulate directly gene expression. [Ca(2+)](N) may change by propagation of [Ca(2+)](C) changes through the nuclear envelope or by direct release of Ca(2+) inside the nucleus. In the last case nuclear and cytosolic signalling can be dissociated. Phosphatidylinositol bisphosphate, phospholipase C and cyclic ADP-ribosyl cyclase are present inside the nucleus. Inositol trisphosphate receptors (IP(3)R) and ryanodine receptors (RyR) have also been found in the nucleus and can be activated by agonists. Furthermore, nuclear location of the synthesizing enzymes and receptors may be atypical, not associated to the nuclear envelope or other membranes. The possible role of nuclear subdomains such as speckles, nucleoplasmic reticulum, multi-macromolecular complexes and nuclear nanovesicles is discussed.
Collapse
Affiliation(s)
- Maria Teresa Alonso
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), c/Sanz y Forés 3, 47003 Valladolid, Spain
| | | |
Collapse
|
14
|
C. Munday D, Hiscox JA, Barr JN. Quantitative proteomic analysis of A549 cells infected with human respiratory syncytial virus subgroup B using SILAC coupled to LC-MS/MS. Proteomics 2010; 10:4320-34. [PMID: 21110324 PMCID: PMC7167978 DOI: 10.1002/pmic.201000228] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 07/01/2010] [Accepted: 08/31/2010] [Indexed: 12/24/2022]
Abstract
Human respiratory syncytial virus (HRSV) is a leading cause of serious lower respiratory tract infections in infants. The virus has two subgroups A and B, which differ in prevalence and (nucleotide) sequence. The interaction of subgroup A viruses with the host cell is relatively well characterized, whereas for subgroup B viruses it is not. Therefore quantitative proteomics was used to investigate the interaction of subgroup B viruses with A549 cells, a respiratory cell line. Changes in the cellular proteome and potential canonical pathways were determined using SILAC coupled to LC-MS/MS and Ingenuity Pathway Analysis. To reduce sample complexity and investigate potential trafficking both nuclear and cytoplasmic fractions were analyzed. A total of 904 cellular and six viral proteins were identified and quantified, of which 112 cellular proteins showed a twofold or more change in HRSV-infected cells. Data sets were validated using indirect immunofluorescence confocal microscopy on independent samples. Major changes were observed in constituents of mitochondria including components of the electron transport chain complexes and channels, as well as increases in the abundance of the products of interferon-stimulated genes. This is the first quantitative proteomic analysis of cells infected with HRSV-subgroup B.
Collapse
Affiliation(s)
- Diane C. Munday
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Julian A. Hiscox
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - John N. Barr
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| |
Collapse
|
15
|
Olins AL, Ernst A, Zwerger M, Herrmann H, Olins DE. An in vitro model for Pelger-Huët anomaly: stable knockdown of lamin B receptor in HL-60 cells. Nucleus 2010; 1:506-12. [PMID: 21327094 PMCID: PMC3027054 DOI: 10.4161/nucl.1.6.13271] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 08/03/2010] [Accepted: 08/06/2010] [Indexed: 11/19/2022] Open
Abstract
The principal human blood granulocyte (neutrophil) possesses a lobulated and deformable nucleus, important to facilitate rapid egress from blood vessels as these cells migrate to sites of bacterial or fungal infection. This unusual nuclear shape is a product of elevated levels of an integral membrane protein of the nuclear envelope lamin B receptor (LBR) and of decreased amounts of lamin A/C. In humans, a genetic deficiency of LBR produces Pelger-Huët anomaly, resulting in blood neutrophils that exhibit hypolobulated nuclei with redistributed heterochromatin. Structural changes in nuclear architecture occur during granulopoiesis within bone marrow. The exact mechanisms of this nuclear shape change and of heterochromatin redistribution remain largely unknown. As a tool to facilitate analysis of these mechanisms, a stable LBR knockdown subline of HL-60 cells was established. During in vitro granulopoiesis induced with retinoic acid, the LBR knockdown cells retain an ovoid shaped nucleus with reduced levels of lamin A/C; while, the parent cells develop highly lobulated nuclei. In contrast, macrophage forms induced in LBR knockdown cells by in vitro treatment with phorbol ester were indistinguishable from the parent cells, judged by both nuclear shape and attached cell morphology. The capability of differentiation of LBR knockdown HL-60 cells should facilitate a detailed analysis of the molecular relationship between LBR levels, granulocyte nuclear shape and heterochromatin distribution.
Collapse
Affiliation(s)
- Ada L Olins
- Department of Biology, Bowdoin College, Brunswick, ME, USA
| | | | | | | | | |
Collapse
|
16
|
Munday DC, Emmott E, Surtees R, Lardeau CH, Wu W, Duprex WP, Dove BK, Barr JN, Hiscox JA. Quantitative proteomic analysis of A549 cells infected with human respiratory syncytial virus. Mol Cell Proteomics 2010; 9:2438-59. [PMID: 20647383 PMCID: PMC2984239 DOI: 10.1074/mcp.m110.001859] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) is a major cause of pediatric lower respiratory tract disease to which there is no vaccine or efficacious chemotherapeutic strategy. Although RNA synthesis and virus assembly occur in the cytoplasm, HRSV is known to induce nuclear responses in the host cell as replication alters global gene expression. Quantitative proteomics was used to take an unbiased overview of the protein changes in transformed human alveolar basal epithelial cells infected with HRSV. Underpinning this was the use of stable isotope labeling with amino acids in cell culture coupled to LC-MS/MS, which allowed the direct and simultaneous identification and quantification of both cellular and viral proteins. To reduce sample complexity and increase data return on potential protein localization, cells were fractionated into nuclear and cytoplasmic extracts. This resulted in the identification of 1,140 cellular proteins and six viral proteins. The proteomics data were analyzed using Ingenuity Pathways Analysis to identify defined canonical pathways and functional groupings. Selected data were validated using Western blot, direct and indirect immunofluorescence confocal microscopy, and functional assays. The study served to validate and expand upon known HRSV-host cell interactions, including those associated with the antiviral response and alterations in subnuclear structures such as the nucleolus and ND10 (promyelocytic leukemia bodies). In addition, novel changes were observed in mitochondrial proteins and functions, cell cycle regulatory molecules, nuclear pore complex proteins and nucleocytoplasmic trafficking proteins. These data shed light into how the cell is potentially altered to create conditions more favorable for infection. Additionally, the study highlights the application and advantage of stable isotope labeling with amino acids in cell culture coupled to LC-MS/MS for the analysis of virus-host interactions.
Collapse
Affiliation(s)
- Diane C Munday
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Dominance of parental genomes in embryonic stem cell/fibroblast hybrid cells depends on the ploidy of the somatic partner. Cell Tissue Res 2010; 340:437-50. [DOI: 10.1007/s00441-010-0987-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2010] [Accepted: 04/21/2010] [Indexed: 11/26/2022]
|
18
|
Bootman MD, Fearnley C, Smyrnias I, MacDonald F, Roderick HL. An update on nuclear calcium signalling. J Cell Sci 2009; 122:2337-50. [PMID: 19571113 DOI: 10.1242/jcs.028100] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Over the past 15 years or so, numerous studies have sought to characterise how nuclear calcium (Ca2+) signals are generated and reversed, and to understand how events that occur in the nucleoplasm influence cellular Ca2+ activity, and vice versa. In this Commentary, we describe mechanisms of nuclear Ca2+ signalling and discuss what is known about the origin and physiological significance of nuclear Ca2+ transients. In particular, we focus on the idea that the nucleus has an autonomous Ca2+ signalling system that can generate its own Ca2+ transients that modulate processes such as gene transcription. We also discuss the role of nuclear pores and the nuclear envelope in controlling ion flux into the nucleoplasm.
Collapse
Affiliation(s)
- Martin D Bootman
- Laboratory of Molecular Signalling, The Babraham Institute, Babraham, Cambridge CB22 3AT, UK.
| | | | | | | | | |
Collapse
|
19
|
Hidalgo C, Donoso P. Crosstalk between calcium and redox signaling: from molecular mechanisms to health implications. Antioxid Redox Signal 2008; 10:1275-312. [PMID: 18377233 DOI: 10.1089/ars.2007.1886] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Studies done many years ago established unequivocally the key role of calcium as a universal second messenger. In contrast, the second messenger roles of reactive oxygen and nitrogen species have emerged only recently. Therefore, their contributions to physiological cell signaling pathways have not yet become universally accepted, and many biological researchers still regard them only as cellular noxious agents. Furthermore, it is becoming increasingly apparent that there are significant interactions between calcium and redox species, and that these interactions modify a variety of proteins that participate in signaling transduction pathways and in other fundamental cellular functions that determine cell life or death. This review article addresses first the central aspects of calcium and redox signaling pathways in animal cells, and continues with the molecular mechanisms that underlie crosstalk between calcium and redox signals under a number of physiological or pathological conditions. To conclude, the review focuses on conditions that, by promoting cellular oxidative stress, lead to the generation of abnormal calcium signals, and how this calcium imbalance may cause a variety of human diseases including, in particular, degenerative diseases of the central nervous system and cardiac pathologies.
Collapse
Affiliation(s)
- Cecilia Hidalgo
- Centro FONDAP de Estudios Moleculares de la Célula and Programa de Biología Molecular y Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | | |
Collapse
|
20
|
Bezin S, Fossier P, Cancela JM. Nucleoplasmic reticulum is not essential in nuclear calcium signalling mediated by cyclic ADPribose in primary neurons. Pflugers Arch 2008; 456:581-6. [PMID: 18197416 DOI: 10.1007/s00424-007-0435-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 12/06/2007] [Accepted: 12/14/2007] [Indexed: 11/30/2022]
Abstract
Nuclear calcium regulation is essential for controlling nuclear processes such as gene expression. Recent studies, mostly performed on immortalized or transformed cell lines, reported the presence of a nucleoplasmic reticulum (NR). It has been suggested that NR acts as a storage organelle having an important role in nuclear Ca2+ signalling. However, whether NR is present and necessary in primary neurons for generation of nuclear Ca2+ signalling has never been investigated. Here, we show, by confocal microscopy and by electronic microscopy, that nuclei in intact neurons or isolated nuclei are not endowed with NR. Finally, our experiments performed on isolated nuclei from Aplysia giant neurons show that the nuclear envelope acts as a functional Ca2+ store which can be mobilized by the second messenger cyclic ADPribose to elicit a nucleoplasmic Ca2+ elevation. Our study provides evidence that nuclear Ca2+ signals can be independent of the presence of NR in neurons.
Collapse
Affiliation(s)
- S Bezin
- CNRS, Institut de Neurobiologie Alfred Fessard-FRC 2118, Laboratoire de Neurobiologie Cellulaire et Moléculaire-UPR9040, 91198 Gif sur Yvette, France
| | | | | |
Collapse
|
21
|
Bussolati G, Marchiò C, Gaetano L, Lupo R, Sapino A. Pleomorphism of the nuclear envelope in breast cancer: a new approach to an old problem. J Cell Mol Med 2007; 12:209-18. [PMID: 18053086 PMCID: PMC3823482 DOI: 10.1111/j.1582-4934.2007.00176.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In routine practice, nuclear pleomorphism of tumours is assessed by haematoxylin staining of the membrane-bound heterochromatin. However, decoration of the nuclear envelope (NE) through the immunofluorescence staining of NE proteins such as lamin B and emerin can provide a more objective appreciation of the nuclear shape. In breast cancer, nuclear pleomorphism is one of the least reproducible parameters to score histological grade, thus we sought to use NE proteins to improve the reproducibility of nuclear grading. First, immuno-fluorescence staining of NE as well as confocal microscopy and three-dimensional reconstruction of nuclei in cultured cells showed a smooth and uniform NE of normal breast epithelium in contrast to an irregular foldings of the membrane and the presence of deep invaginations leading to the formation of an intranuclear scaffold of NE-bound tubules in breast cancer cells. Following the above methods and criteria, we recorded the degree of NE pleomorphism (NEP) in a series of 273 invasive breast cancers tested by immunofluorescence. A uniform nuclear shape with few irregularities (low NEP) was observed in 135 cases or, alternatively, marked folds of the NE and an intranuclear tubular scaffold (high NEP cases) were observed in 138 cases. The latter features were significantly correlated (P-value <0.002) with lymph node metastases in 54 histological grade 1 and in 173 cancers with low mitotic count. Decoration of the NE might thus be regarded as a novel diagnostic parameter to define the grade of malignancy, which parallels and enhances that provided by routine histological procedures.
Collapse
Affiliation(s)
- Gianni Bussolati
- Department of Biomedical Science and Human Oncology, University of Torino, Via Santena, Torino, Italy.
| | | | | | | | | |
Collapse
|
22
|
Chamero P, Manjarres IM, García-Verdugo JM, Villalobos C, Alonso MT, García-Sancho J. Nuclear calcium signaling by inositol trisphosphate in GH3 pituitary cells. Cell Calcium 2007; 43:205-14. [PMID: 17583789 DOI: 10.1016/j.ceca.2007.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2007] [Revised: 05/01/2007] [Accepted: 05/08/2007] [Indexed: 11/28/2022]
Abstract
It has been proposed that nuclear and cytosolic Ca(2+) ([Ca(2+)](N) and [Ca(2+)](C)) may be regulated independently. We address here the issue of whether inositol trisphosphate (IP(3)) can, bypassing changes of [Ca(2+)](C), produce direct release of Ca(2+) into the nucleoplasm. We have used targeted aequorins to selectively measure and compare the changes in [Ca(2+)](C) and [Ca(2+)](N) induced by IP(3) in GH(3) pituitary cells. Heparin, an IP(3) inhibitor that does not permeate the nuclear pores, abolished the [Ca(2+)](C) peaks but inhibited only partly the [Ca(2+)](N) peaks. The permeant inhibitor 2-aminoethoxy-diphenyl-borate (2-APB) blocked both responses. Removal of ATP also inhibited more strongly the [Ca(2+)](C) than [Ca(2+)](N) peak. The [Ca(2+)](N) and [Ca(2+)](C) responses differed also in their sensitivity to IP(3), the nuclear response showing higher affinity. Among IP(3) receptors, type 2 (IP(3)R2) has a higher affinity for IP(3) and is not inactivated by ATP removal. We find that IP(3)R2 immunoreactivity is present inside the nucleus whereas the other IP(3)R subtypes are detected only in the cytoplasm. The nuclear envelope (NE) of GH(3) cells showed deep invaginations into the nucleoplasm, with cytosol and cytoplasmic organella inside. These results indicate that GH(3) pituitary cells possess mechanisms able to produce selective increases of [Ca(2+)](N).
Collapse
Affiliation(s)
- Pablo Chamero
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Departamento de Fisiología y Bioquímica, Facultad de Medicina, E-47005 Valladolid, Spain
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Calcium plays an integral role in cellular function. It is a well-recognized second messenger necessary for signaling cellular responses, but in excessive amounts can be deleterious to function, causing cell death. The main route by which calcium enters the cytoplasm is either from the extracellular compartment or internal addistores via calcium channels. There is good evidence that calcium channels can respond to pharmacological compounds that reduce or oxidize thiol groups on the channel protein. In addition, reactive oxygen species such as hydrogen peroxide and superoxide that can mediate oxidative pathology also mediate changes in channel function via alterations of thiol groups. This review looks at the structure and function of calcium channels, the evidence that changes in cellular redox state mediate changes in channel function, and the role of redox modification of channels in disease processes. Understanding how redox modification of the channel protein alters channel structure and function is providing leads for the design of therapeutic interventions that target oxidative stress responses.
Collapse
Affiliation(s)
- Livia C Hool
- Discipline of Physiology, School of Biomedical, Biomolecular, and Chemical Sciences, The University of Western Australia, Crawley, Western Australia.
| | | |
Collapse
|
24
|
George CH, Rogers SA, Bertrand BMA, Tunwell REA, Thomas NL, Steele DS, Cox EV, Pepper C, Hazeel CJ, Claycomb WC, Lai FA. Alternative Splicing of Ryanodine Receptors Modulates Cardiomyocyte Ca
2+
Signaling and Susceptibility to Apoptosis. Circ Res 2007; 100:874-83. [PMID: 17322175 DOI: 10.1161/01.res.0000260804.77807.cf] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ca
2+
release via type 2 ryanodine receptors (RyR2) regulates cardiac function. Molecular cloning of human RyR2 identified 2 alternatively spliced variants, comprising 30- and 24-bp sequence insertions; yet their role in shaping cardiomyocyte Ca
2+
signaling and cell phenotype is unknown. We profiled the developmental regulation and the tissue and species specificity of these variants and showed that their recombinant expression in HL-1 cardiomyocytes profoundly modulated nuclear and cytoplasmic Ca
2+
release. All splice variants localized to the sarcoplasmic reticulum, perinuclear Golgi apparatus, and to finger-like invaginations of the nuclear envelope (nucleoplasmic reticulum). Strikingly, the 24-bp splice insertion that was present at low levels in embryonic and adult hearts was essential for targeting RyR2 to an intranuclear Golgi apparatus and promoted the intracellular segregation of this variant. The amplitude variability of nuclear and cytoplasmic Ca
2+
fluxes were reduced in nonstimulated cardiomyocytes expressing both 30- and 24-bp splice variants and were associated with lower basal levels of apoptosis. Expression of RyR2 containing the 24-bp insertion also suppressed intracellular Ca
2+
fluxes following prolonged caffeine exposure (1 mmol/L, 16 hours) that protected cells from apoptosis. The antiapoptotic effects of this variant were linked to increased levels of Bcl-2 phosphorylation. In contrast, RyR2 containing the 30-bp insertion, which was abundant in human embryonic heart but was decreased during cardiac development, did not protect cardiomyocytes from caffeine-evoked apoptosis. Thus, we provide the first evidence that RyR2 splice variants exquisitely modulate intracellular Ca
2+
signaling and are key determinants of cardiomyocyte apoptotic susceptibility.
Collapse
Affiliation(s)
- Christopher H George
- Department of Cardiology, Wales Heart Research Institute, School of Medicine, Cardiff University, Heath Park, Cardiff, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Alonso MT, Villalobos C, Chamero P, Alvarez J, García-Sancho J. Calcium microdomains in mitochondria and nucleus. Cell Calcium 2006; 40:513-25. [PMID: 17067669 DOI: 10.1016/j.ceca.2006.08.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Accepted: 08/23/2006] [Indexed: 10/24/2022]
Abstract
Endomembranes modify the progression of the cytosolic Ca(2+) wave and contribute to generate Ca(2+) microdomains, both in the cytosol and inside the own organella. The concentration of Ca(2+) in the cytosol ([Ca(2+)](C)), the mitochondria ([Ca(2+)](M)) and the nucleus ([Ca(2+)](N)) are similar at rest, but may become very different during cell activation. Mitochondria avidly take up Ca(2+) from the high [Ca(2+)](C) microdomains generated during cell activation near Ca(2+) channels of the plasma membrane and/or the endomembranes and prevent propagation of the high Ca(2+) signal to the bulk cytosol. This shaping of [Ca(2+)](C) signaling is essential for independent regulation of compartmentalized cell functions. On the other hand, a high [Ca(2+)](M) signal is generated selectively in the mitochondria close to the active areas, which tunes up respiration to the increased local needs. The progression of the [Ca(2+)](C) signal to the nucleus may be dampened by mitochondria, the nuclear envelope or higher buffering power inside the nucleoplasm. On the other hand, selective [Ca(2+)](N) signals could be generated by direct release of stored Ca(2+) into the nucleoplasm. Ca(2+) release could even be restricted to subnuclear domains. Putative Ca(2+) stores include the nuclear envelope, their invaginations inside the nucleoplasm (nucleoplasmic reticulum) and nuclear microvesicles. Inositol trisphosphate, cyclic ADP-ribose and nicotinic acid adenine dinucleotide phosphate have all been reported to produce release of Ca(2+) into the nucleoplasm, but contribution of these mechanisms under physiological conditions is still uncertain.
Collapse
Affiliation(s)
- María Teresa Alonso
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), c/Sanz y Forés s/n, Valladolid, Spain
| | | | | | | | | |
Collapse
|
26
|
Abstract
Lipids are not only components of cell nucleus membranes, but are also found in the membrane-depleted nuclei where they fulfill special functions. We have investigated the lipid composition of membrane-depleted rat liver nuclei obtained by incubation with low Triton X-100 concentrations of 0.04% and 0.08%, which rendered them unaltered or hardly altered. Under these conditions, 26% of proteins and 22% of phospholipids were recovered. The main phospholipids were phosphatidylcholine > phosphatidylethanolamine > phosphatidylinositol = or > phosphatidylserine and sphingomyelin (in decreasing concentrations). The fatty acid components of total lipids and phosphatidylcholine were mainly unsaturated. Over 40% belonged to the n-6 series (arachidonic > or = 25% and linoleic 15%); approximately 40% corresponded to saturated acids and <10% were monoenoic. Endonuclear phosphatidylcholine was built up by 16 molecular species, the most abundant being 18:0-20:4 (32%), 16:0-20:4 (19%), 16:0-18:2 (13%), and 18:0-18:2 (11%). The fatty acid composition and phosphatidylcholine molecular species distribution in the membrane-depleted nucleus of rat liver showed patterns similar to the whole nucleus, mitochondria, microsomes, and homogenate of the parent liver cells, suggesting that endonuclear lipid pool composition is mainly determined by a liver organ profile.
Collapse
Affiliation(s)
- Sabina M Maté
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Facultad de Ciencias Médicas, UNLP-CONICET, calles 60 y 120, Argentina
| | | | | |
Collapse
|
27
|
Underwood JM, Imbalzano KM, Weaver VM, Fischer AH, Imbalzano AN, Nickerson JA. The ultrastructure of MCF-10A acini. J Cell Physiol 2006; 208:141-8. [PMID: 16607610 DOI: 10.1002/jcp.20639] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
MCF-10A human mammary epithelial cells cultured inside reconstituted basement membrane form acini that resemble the acinar structures of mammary lobules. This three-dimensional culture system has been used for identifying and characterizing the signal transduction pathways controlling cell proliferation and death, and for studying their disregulation in malignant progression. We have compared the ultrastructure of MCF-10A acini, MCF-10A cells grown in monolayer, and the acinar structures of human breast lobules. The tissue architecture of MCF-10A acini was formed by hemidesmosomes connected to a basement membrane and by abundant desmosomes between acinar cells. Intermediate filaments that joined into large and abundant filament bundles connected hemidesmosomes and desmosomes to sites at the nuclear surface. Fewer and thinner bundles of filaments were observed in monolayer MCF-10A cells and even fewer in breast tissue. Tight junctions were observed between cells in breast tissue but missing in MCF-10A acini. The cytoplasm of MCF-10A acinar cells had a polar organization similar to that observed in breast tissue, with centrosomes and the Golgi apparatus on the apical side of the nucleus. MCF-10A acinar nuclei had an irregular, frequently invaginated surface and had a single nucleolus. The distribution of heterochromatin was similar to that in the epithelial cells of breast tissue. The nuclei of monolayer MCF-10A cells had multiple nucleoli, a more regular profile, and less heterochromatin. Electron microscopy has the resolution required to survey features of MCF-10A cell and acinus architecture that may change with manipulations designed to induce malignant phenotypes.
Collapse
Affiliation(s)
- Jean M Underwood
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | |
Collapse
|
28
|
Lee RKY, Lui PPY, Ngan EKS, Lui JCK, Suen YK, Chan F, Kong SK. The nuclear tubular invaginations are dynamic structures inside the nucleus of HeLa cellsThis paper is one of a selection of papers published in this Special Issue, entitled The Nucleus: A Cell Within A Cell. Can J Physiol Pharmacol 2006; 84:477-86. [PMID: 16902593 DOI: 10.1139/y05-110] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nuclear tubules (NTs) were found in the nucleus of HeLa cells. Although no function has been ascribed to these structures, our previous data has shown that they are the sites of Ca2+release with mitochondria shuttled around. In the present study, we further characterized these NTs through different fluorescent dye-labeling and red fluorescent protein transfection experiments. We found that doxorubicin (Dox) is a good indicator to demonstrate the NTs since Dox is fluorescent and DNA is able to quench its fluorescence. By using confocal and electron microscopy, we show that the number and nature of the NTs in HeLa vary from cell to cell, ranging from tubular to intricately branched structures. Additionally, these NTs are double-membrane invaginations of the nuclear envelope and usually lie close to nucleolus. At rest, NTs appeared to be stable and their mouths are always closed. Upon Ca2+ionomycin stimulation, various forms of dynamism, including membrane protrusion to the nucleus, enlargement and shrinkage of the NTs, and distortion of the nuclear envelope and NTs were observed over a time scale of minutes. These observations suggest that the NT represents a specialized and dynamic compartment inside the nucleus under the control of Ca2+.
Collapse
Affiliation(s)
- Rebecca K Y Lee
- Department of Biochemistry, Basic Medical Sciences Building, The Chinese University of Hong Kong, Shatin, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Cárdenas C, Liberona JL, Molgó J, Colasante C, Mignery GA, Jaimovich E. Nuclear inositol 1,4,5-trisphosphate receptors regulate local Ca2+ transients and modulate cAMP response element binding protein phosphorylation. J Cell Sci 2006; 118:3131-40. [PMID: 16014380 DOI: 10.1242/jcs.02446] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several lines of evidence indicate that increases in nuclear Ca(2+) have specific biological effects that differ from those of cytosolic Ca(2+), suggesting that they occur independently. The mechanisms involved in controlling nuclear Ca(2+) signaling are both controversial and still poorly understood. Using hypotonic shock combined with mechanical disruption, we obtained and characterized a fraction of purified nuclei from cultured rat skeletal myotubes. Both immunoblot studies and radiolabeled inositol 1,4,5-trisphosphate [IP(3)] binding revealed an important concentration of IP(3) receptors in the nuclear fraction. Immunofluorescence and immunoelectron microscopy studies localized type-1 and type-3 IP(3) receptors in the nucleus with type-1 receptors preferentially localized in the inner nuclear membrane. Type-2 IP(3) receptor was confined to the sarcoplasmic reticulum. Isolated nuclei responded to IP(3) with rapid and transient Ca(2+) concentration elevations, which were inhibited by known blockers of IP(3) signals. Similar results were obtained with isolated nuclei from the 1B5 cell line, which does not express ryanodine receptors but releases nuclear Ca(2+) in an IP(3)-dependent manner. Nuclear Ca(2+) increases triggered by IP(3) evoked phosphorylation of cAMP response element binding protein with kinetics compatible with sequential activation. These results support the idea that Ca(2+) signals, mediated by nuclear IP(3) receptors in muscle cells, are part of a distinct Ca(2+) release component that originates in the nucleus and probably participates in gene regulation mediated by cAMP response element binding protein.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Calcium/metabolism
- Calcium Channels/biosynthesis
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/physiology
- Cell Nucleus/metabolism
- Cells, Cultured
- Cyclic AMP Response Element-Binding Protein/metabolism
- Fluorometry
- Immunohistochemistry
- Inositol 1,4,5-Trisphosphate/metabolism
- Inositol 1,4,5-Trisphosphate Receptors
- Microscopy, Confocal
- Microscopy, Immunoelectron
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Nuclear Envelope/metabolism
- Phosphorylation
- Protein Binding
- Protein Isoforms
- Rats
- Receptors, Cytoplasmic and Nuclear/biosynthesis
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
Collapse
Affiliation(s)
- Cesar Cárdenas
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 7, Chile
| | | | | | | | | | | |
Collapse
|
30
|
Koopman WJH, Willems PHGM, Oosterhof A, van Kuppevelt TH, Gielen SCAM. Amplitude modulation of nuclear Ca2+ signals in human skeletal myotubes: A possible role for nuclear Ca2+ buffering. Cell Calcium 2005; 38:141-52. [PMID: 16054687 DOI: 10.1016/j.ceca.2005.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Revised: 05/17/2005] [Accepted: 06/02/2005] [Indexed: 11/20/2022]
Abstract
Video-rate confocal microscopy of Indo-1-loaded human skeletal myotubes was used to assess the relationship between the changes in sarcoplasmic ([Ca(2+)](S)) and nuclear ([Ca(2+)](N)) Ca(2+) concentration during low- and high-frequency electrostimulation. A single stimulus of 10 ms duration transiently increased [Ca(2+)] in both compartments with the same time of onset. Rate and amplitude of the [Ca(2+)] rise were significantly lower in the nucleus (4.0- and 2.5-fold, respectively). Similarly, [Ca(2+)](N) decayed more slowly than [Ca(2+)](S) (mono-exponential time constants of 6.1 and 2.5 s, respectively). After return of [Ca(2+)] to the prestimulatory level, a train of 10 stimuli was applied at a frequency of 1 Hz. The amplitude of the first [Ca(2+)](S) transient was 25% lower than that of the preceding single transient. Thereafter, [Ca(2+)](S) increased stepwise to a maximum that equalled that of the single transient. Similarly, the amplitude of the first [Ca(2+)](N) transient was 20% lower than that of the preceding single transient. In contrast to [Ca(2+)](S), [Ca(2+)](N) then increased to a maximum that was 2.3-fold higher than that of the single transient and equalled that of [Ca(2+)](S). In the nucleus, and to a lesser extent in the sarcoplasm, [Ca(2+)] decreased faster at the end of the stimulus train than after the preceding single stimulus (time constants of 3.3 and 2.1 s, respectively). To gain insight into the molecular principles underlying the shaping of the nuclear Ca(2+) signal, a 3-D mathematical model was constructed. Intriguingly, quantitative modelling required the inclusion of a satiable nuclear Ca(2+) buffer. Alterations in the concentration of this putative buffer had dramatic effects on the kinetics of the nuclear Ca(2+) signal. This finding unveils a possible mechanism by which the skeletal muscle can adapt to changes in physiological demand.
Collapse
Affiliation(s)
- Werner J H Koopman
- Department of 160 Biochemistry NCMLS, Radboud University Nijmegen Medical Center, The Netherlands
| | | | | | | | | |
Collapse
|
31
|
Florea AM, Yamoah EN, Dopp E. Intracellular calcium disturbances induced by arsenic and its methylated derivatives in relation to genomic damage and apoptosis induction. ENVIRONMENTAL HEALTH PERSPECTIVES 2005; 113:659-64. [PMID: 15929885 PMCID: PMC1257587 DOI: 10.1289/ehp.7634] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Arsenic and its methylated derivatives are contaminants of air, water, and food and are known as toxicants and carcinogens. Arsenic compounds are also being used as cancer chemotherapeutic agents. In humans, inorganic arsenic is metabolically methylated to mono-, di-, and trimethylated forms. Recent findings suggest that the methylation reactions represent a toxification rather than a detoxification pathway. In recent years, the correlation between arsenic exposure, cytotoxicity and genotoxicity, mutagenicity, and tumor promotion has been established, as well as the association of arsenic exposure with perturbation of physiologic processes, generation of reactive oxygen species, DNA damage, and apoptosis induction. Trivalent forms of arsenic have been found to induce apoptosis in several cellular systems with involvement of membrane-bound cell death receptors, activation of caspases, release of calcium stores, and changes of the intracellular glutathione level. It is well known that calcium ion deregulation plays a critical role in apoptotic cell death. A calcium increase in the nuclei might lead to toxic effects in the cell. In this review, we highlight the relationship between induced disturbances of calcium homeostasis, genomic damage, and apoptotic cell death caused by arsenic and its organic derivatives.
Collapse
Affiliation(s)
- Ana-Maria Florea
- Institute of Hygiene and Occupational Medicine, University Hospital, Essen, Germany
| | | | | |
Collapse
|
32
|
Abstract
The endoplasmic reticulum is not the only major agonist-releasable Ca2+ store within cells; it is now clear that virtually all organelles so far studied have the ability to act as mobilizable Ca2+ stores. From recent findings with regard to Ca2+ transportation and Ca2+ homeostasis within a variety of cell organelles such as the mitochondria, nucleus, Golgi and lysosomes, it emerges that many of these organellar Ca2+ stores appear to interact with each other, adding a further level of complexity to Ca2+ signalling events.
Collapse
|
33
|
Florea AM, Dopp E, Büsselberg D. Elevated Ca2+i transients induced by trimethyltin chloride in HeLa cells: types and levels of response. Cell Calcium 2005; 37:251-8. [PMID: 15670872 DOI: 10.1016/j.ceca.2004.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2004] [Revised: 10/07/2004] [Accepted: 10/26/2004] [Indexed: 11/26/2022]
Abstract
Humans are exposed to organotins, like trimethyltin (TMT) chloride via air, water and food, and intoxication might result in severe health complications. Toxic effects of organotin compounds are well documented, but possible mechanisms remain unclear and only little information is available how organometallic species interact with calcium controlling mechanisms. Therefore, the aim of this work was to investigate the effects of TMT on calcium homeostasis in HeLa S3 cells. Dynamic changes of cytosolic calcium (Ca2+(i)) were monitored using laser-scanning microscopy and fluo-4 loaded cells. Application of TMT resulted in sustained as well as in transient elevations of Ca2+(i). The number of reacting cells was directly correlated to the concentration of TMT used: with 500 microM TMT all cells reacted, with 50 microM TMT 80% and with 5 microM 74%. The fast Ca2+(i)-transients (spikes), measured in single cells, occurred even with 0.25 microM TMT and varied in size and duration. The sustained increase of Ca2+(i), measured as the average over all cells, was dose dependent with an approximately 8% increase for 5 microM TMT, approximately 12.3% for 50 microM and approximately 145% for 500 microM TMT. Moreover, this effect was partly reversible. A second application resulted in a similar sustained rise of Ca2+(i) compared to the first application of TMT, there was also no difference when no calcium was added to the external solution (151+/-10% compared to 145+/-15%; 500 microM TMT). This rise of Ca2+(i) was highly reduced (<10% increase) when the internal calcium stores were depleted before TMT (500 microM) was applied. Our data suggest that TMT influences Ca2+(i)-homeostasis of HeLa S3 cells, which might be related to its toxicity in this cell line.
Collapse
Affiliation(s)
- Ana-Maria Florea
- Institute of Hygiene and Occupational Medicine, University Hospital Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | |
Collapse
|