1
|
Li Q, Fang J, Liu K, Luo P, Wang X. Multi-omic validation of the cuproptosis-sphingolipid metabolism network: modulating the immune landscape in osteosarcoma. Front Immunol 2024; 15:1424806. [PMID: 38983852 PMCID: PMC11231095 DOI: 10.3389/fimmu.2024.1424806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Background The current understanding of the mechanisms by which metal ion metabolism promotes the progression and drug resistance of osteosarcoma remains incomplete. This study aims to elucidate the key roles and mechanisms of genes involved in cuproptosis-related sphingolipid metabolism (cuproptosis-SPGs) in regulating the immune landscape, tumor metastasis, and drug resistance in osteosarcoma cells. Methods This study employed multi-omics approaches to assess the impact of cuproptosis-SPGs on the prognosis of osteosarcoma patients. Lasso regression analysis was utilized to construct a prognostic model, while multivariate regression analysis was applied to identify key core genes and generate risk coefficients for these genes, thereby calculating a risk score for each osteosarcoma patient. Patients were then stratified into high-risk and low-risk groups based on their risk scores. The ESTIMATE and CIBERSORT algorithms were used to analyze the level of immune cell infiltration within these risk groups to construct the immune landscape. Single-cell analysis was conducted to provide a more precise depiction of the expression patterns of cuproptosis-SPGs among immune cell subtypes. Finally, experiments on osteosarcoma cells were performed to validate the role of the cuproptosis-sphingolipid signaling network in regulating cell migration and apoptosis. Results In this study, seven cuproptosis-SPGs were identified and used to construct a prognostic model for osteosarcoma patients. In addition to predicting survival, the model also demonstrated reliability in forecasting the response to chemotherapy drugs. The results showed that a high cuproptosis-sphingolipid metabolism score was closely associated with reduced CD8 T cell infiltration and indicated poor prognosis in osteosarcoma patients. Cellular functional assays revealed that cuproptosis-SPGs regulated the LC3B/ERK signaling pathway, thereby triggering cell death and impairing migration capabilities in osteosarcoma cells. Conclusion The impact of cuproptosis-related sphingolipid metabolism on the survival and migration of osteosarcoma cells, as well as on CD8 T cell infiltration, highlights the potential of targeting copper ion metabolism as a promising strategy for osteosarcoma patients.
Collapse
Affiliation(s)
- Qingbiao Li
- Department of Orthopedics, Southern Medical University Pingshan Hospital (Pingshan District Peoples’ Hospital of Shenzhen), Shenzhen, Guangdong, China
| | - Jiarui Fang
- Department of Sport Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Kai Liu
- Department of Sport Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Peng Luo
- Department of Sport Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Xiuzhuo Wang
- Department of Orthopedics, Southern Medical University Pingshan Hospital (Pingshan District Peoples’ Hospital of Shenzhen), Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Gulbins A, Görtz GE, Gulbins E, Eckstein A. Sphingolipids in thyroid eye disease. Front Endocrinol (Lausanne) 2023; 14:1170884. [PMID: 37082124 PMCID: PMC10112667 DOI: 10.3389/fendo.2023.1170884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/22/2023] [Indexed: 04/22/2023] Open
Abstract
Graves' disease (GD) is caused by an autoimmune formation of autoantibodies and autoreactive T-cells against the thyroid stimulating hormone receptor (TSHR). The autoimmune reaction does not only lead to overstimulation of the thyroid gland, but very often also to an immune reaction against antigens within the orbital tissue leading to thyroid eye disease, which is characterized by activation of orbital fibroblasts, orbital generation of adipocytes and myofibroblasts and increased hyaluronan production in the orbit. Thyroid eye disease is the most common extra-thyroidal manifestation of the autoimmune Graves' disease. Several studies indicate an important role of sphingolipids, in particular the acid sphingomyelinase/ceramide system and sphingosine 1-phosphate in thyroid eye disease. Here, we discuss how the biophysical properties of sphingolipids contribute to cell signaling, in particular in the context of thyroid eye disease. We further review the role of the acid sphingomyelinase/ceramide system in autoimmune diseases and its function in T lymphocytes to provide some novel hypotheses for the pathogenesis of thyroid eye disease and potentially allowing the development of novel treatments.
Collapse
Affiliation(s)
- Anne Gulbins
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Gina-Eva Görtz
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Anja Eckstein, ; Erich Gulbins,
| | - Anja Eckstein
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Anja Eckstein, ; Erich Gulbins,
| |
Collapse
|
3
|
Sekunov AV, Protopopov VA, Skurygin VV, Shalagina MN, Bryndina IG. Muscle Plasticity under Functional Unloading: Effects of an Acid Sphingomyelinase Inhibitor Clomipramine. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
4
|
Jaddoa E, Masania J, Masiero E, Sgamma T, Arroo R, Sillence D, Zetterström T. Effect of antidepressant drugs on the brain sphingolipid system. J Psychopharmacol 2020; 34:716-725. [PMID: 32403969 DOI: 10.1177/0269881120915412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Major depression is a common mood disorder and the central sphingolipid system has been identified as a possible drug target of this condition. Here we investigated the action of antidepressant drugs on sphingolipid levels in rat brain regions, plasma and in cultured mouse macrophages. METHODS Two antidepressant drugs were tested: the serotonin reuptake inhibitor paroxetine and the noradrenaline reuptake inhibitor desipramine, either following acute or chronic treatments. Content of sphingosine and ceramide were analysed using LC-MS or HPLC-UV, respectively. This was from samples of brain, plasma and cultured mouse macrophages. Antidepressant-induced effects on mRNA expression for two key genes of the sphingolipid pathway, SMPD1 and ASAH1, were also measured by using quantitative real-time PCR. RESULTS Chronic but not acute administration of paroxetine or desipramine reduced sphingosine levels in the prefrontal cortex and hippocampus (only paroxetine) but not in the striatum. Ceramide levels were also measured in the hippocampus following chronic paroxetine and likewise to sphingosine this treatment reduced its levels. The corresponding collected plasma samples from chronically treated animals did not show any decrease of sphingosine compared to the corresponding controls. Both drugs failed to reduce sphingosine levels from cultured mouse macrophages. The drug-induced decrease of sphingolipids coincided with reduced mRNA expression of two enzymes of the central sphingolipid pathway, i.e. acid sphingomyelinase (SMPD1) and acid ceramidase (ASAH1). CONCLUSIONS This study supports the involvement of brain sphingolipids in the mechanism of action by antidepressant drugs and for the first time highlights their differential effects on brain versus plasma levels.
Collapse
Affiliation(s)
- Estabraq Jaddoa
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Jinit Masania
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Eva Masiero
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Tiziana Sgamma
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Randolph Arroo
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Daniel Sillence
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Tyra Zetterström
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| |
Collapse
|
5
|
Vaquer CC, Suhaiman L, Pavarotti MA, De Blas GA, Belmonte SA. Ceramide induces a multicomponent intracellular calcium increase triggering the acrosome secretion in human sperm. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118704. [PMID: 32194132 DOI: 10.1016/j.bbamcr.2020.118704] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
Exocytosis of spermatozoon's secretory vesicle, named acrosome reaction (AR), is a regulated event that plays a central role in fertilization. It is coupled to a complex calcium signaling. Ceramide is a multitasking lipid involved in exocytosis. Nevertheless, its effect on secretion is controversial and the underlying cellular and molecular mechanisms remain unknown. Human spermatozoa are useful to dissect the role of ceramide in secretion given that the gamete is not capable to undergo any trafficking mechanisms other than exocytosis. We report for the first time, the presence of sphingolipid metabolism enzymes such as neutral-sphingomyelinase and ceramide synthase in sperm. Ceramidases are also present and active. Both the addition of cell-permeable ceramide and the rise of the endogenous one, increase intracellular calcium acting as potent inducers of exocytosis. Ceramide triggers AR in capacitated spermatozoa and enhances the gamete response to progesterone. The lipid induces physiological ultrastructural changes in the acrosome and triggers an exocytosis-signaling cascade involving protein tyrosine phosphatase 1B and VAMP2. Real-time imaging showed an increment of calcium in the cytosol upon ceramide treatment either in the absence or in the presence of extracellular calcium. Pharmacological experiments demonstrate that at early stages the process involves ryanodine receptors, CatSper (calcium channel of sperm), and store-operated calcium channels. We set out the signaling sequence of events that connect ceramide to internal calcium mobilization and external calcium signals during secretion. These results allow the coordination of lipids and proteins in a pathway that accomplishes secretion. Our findings contribute to the understanding of ceramide's role in regulated exocytosis and fertilization.
Collapse
Affiliation(s)
- Cintia Celina Vaquer
- Instituto de Histología y Embriología de Mendoza (IHEM) "Dr. Mario H. Burgos", CONICET, Universidad Nacional de Cuyo, 5500 Mendoza, Argentina
| | - Laila Suhaiman
- Instituto Interdisciplinario de Ciencias Básicas (ICB), CONICET, Universidad Nacional de Cuyo, 5500 Mendoza, Argentina
| | - Martín Alejandro Pavarotti
- Instituto de Histología y Embriología de Mendoza (IHEM) "Dr. Mario H. Burgos", CONICET, Universidad Nacional de Cuyo, 5500 Mendoza, Argentina
| | - Gerardo Andrés De Blas
- Instituto de Histología y Embriología de Mendoza (IHEM) "Dr. Mario H. Burgos", CONICET, Universidad Nacional de Cuyo, 5500 Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Silvia Alejandra Belmonte
- Instituto de Histología y Embriología de Mendoza (IHEM) "Dr. Mario H. Burgos", CONICET, Universidad Nacional de Cuyo, 5500 Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
6
|
Huang S, Che J, Chu Q, Zhang P. The Role of NLRP3 Inflammasome in Radiation-Induced Cardiovascular Injury. Front Cell Dev Biol 2020; 8:140. [PMID: 32226786 PMCID: PMC7080656 DOI: 10.3389/fcell.2020.00140] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The increasing risk of long-term adverse effects from radiotherapy on the cardiovascular structure is receiving increasing attention. However, the mechanisms underlying this increased risk remain poorly understood. Recently, the nucleotide-binding domain and leucine-rich-repeat-containing family pyrin 3 (NLRP3) inflammasome was suggested to play a critical role in radiation-induced cardiovascular injury. However, the relationship between ionizing radiation and the NLRP3 inflammasome in acute and chronic inflammation is complex. We reviewed literature detailing pathological changes and molecular mechanisms associated with radiation-induced damage to the cardiovascular structure, with a specific focus on NLRP3 inflammasome-related cardiovascular diseases. We also summarized possible therapeutic strategies for the prevention of radiation-induced heart disease (RIHD).
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Che
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Avota E, de Lira MN, Schneider-Schaulies S. Sphingomyelin Breakdown in T Cells: Role of Membrane Compartmentalization in T Cell Signaling and Interference by a Pathogen. Front Cell Dev Biol 2019; 7:152. [PMID: 31457008 PMCID: PMC6700246 DOI: 10.3389/fcell.2019.00152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022] Open
Abstract
Sphingolipids are major components of cellular membranes, and at steady-state level, their metabolic fluxes are tightly controlled. On challenge by external signals, they undergo rapid turnover, which substantially affects the biophysical properties of membrane lipid and protein compartments and, consequently, signaling and morphodynamics. In T cells, external cues translate into formation of membrane microdomains where proximal signaling platforms essential for metabolic reprograming and cytoskeletal reorganization are organized. This review will focus on sphingomyelinases, which mediate sphingomyelin breakdown and ensuing ceramide release that have been implicated in T-cell viability and function. Acting at the sphingomyelin pool at the extrafacial or cytosolic leaflet of cellular membranes, acid and neutral sphingomyelinases organize ceramide-enriched membrane microdomains that regulate T-cell homeostatic activity and, upon stimulation, compartmentalize receptors, membrane proximal signaling complexes, and cytoskeletal dynamics as essential for initiating T-cell motility and interaction with endothelia and antigen-presenting cells. Prominent examples to be discussed in this review include death receptor family members, integrins, CD3, and CD28 and their associated signalosomes. Progress made with regard to experimental tools has greatly aided our understanding of the role of bioactive sphingolipids in T-cell biology at a molecular level and of targets explored by a model pathogen (measles virus) to specifically interfere with their physiological activity.
Collapse
Affiliation(s)
- Elita Avota
- Institute for Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Maria Nathalia de Lira
- Institute for Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
8
|
Pulli I, Asghar MY, Kemppainen K, Törnquist K. Sphingolipid-mediated calcium signaling and its pathological effects. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1668-1677. [DOI: 10.1016/j.bbamcr.2018.04.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/17/2018] [Accepted: 04/23/2018] [Indexed: 12/15/2022]
|
9
|
Becker KA, Riethmüller J, Seitz AP, Gardner A, Boudreau R, Kamler M, Kleuser B, Schuchman E, Caldwell CC, Edwards MJ, Grassmé H, Brodlie M, Gulbins E. Sphingolipids as targets for inhalation treatment of cystic fibrosis. Adv Drug Deliv Rev 2018; 133:66-75. [PMID: 29698625 DOI: 10.1016/j.addr.2018.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 01/19/2023]
Abstract
Studies over the past several years have demonstrated the important role of sphingolipids in cystic fibrosis (CF), chronic obstructive pulmonary disease and acute lung injury. Ceramide is increased in airway epithelial cells and alveolar macrophages of CF mice and humans, while sphingosine is dramatically decreased. This increase in ceramide results in chronic inflammation, increased death of epithelial cells, release of DNA into the bronchial lumen and thereby an impairment of mucociliary clearance; while the lack of sphingosine in airway epithelial cells causes high infection susceptibility in CF mice and possibly patients. The increase in ceramide mediates an ectopic expression of β1-integrins in the luminal membrane of CF epithelial cells, which results, via an unknown mechanism, in a down-regulation of acid ceramidase. It is predominantly this down-regulation of acid ceramidase that results in the imbalance of ceramide and sphingosine in CF cells. Correction of ceramide and sphingosine levels can be achieved by inhalation of functional acid sphingomyelinase inhibitors, recombinant acid ceramidase or by normalization of β1-integrin expression and subsequent re-expression of endogenous acid ceramidase. These treatments correct pulmonary inflammation and prevent or treat, respectively, acute and chronic pulmonary infections in CF mice with Staphylococcus aureus and mucoid or non-mucoid Pseudomonas aeruginosa. Inhalation of sphingosine corrects sphingosine levels only and seems to mainly act against the infection. Many antidepressants are functional inhibitors of the acid sphingomyelinase and were designed for systemic treatment of major depression. These drugs could be repurposed to treat CF by inhalation.
Collapse
|
10
|
Börtlein C, Draeger A, Schoenauer R, Kuhlemann A, Sauer M, Schneider-Schaulies S, Avota E. The Neutral Sphingomyelinase 2 Is Required to Polarize and Sustain T Cell Receptor Signaling. Front Immunol 2018; 9:815. [PMID: 29720981 PMCID: PMC5915489 DOI: 10.3389/fimmu.2018.00815] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/04/2018] [Indexed: 01/02/2023] Open
Abstract
By promoting ceramide release at the cytosolic membrane leaflet, the neutral sphingomyelinase 2 (NSM) is capable of organizing receptor and signalosome segregation. Its role in T cell receptor (TCR) signaling remained so far unknown. We now show that TCR-driven NSM activation is dispensable for TCR clustering and initial phosphorylation, but of crucial importance for further signal amplification. In particular, at low doses of TCR stimulatory antibodies, NSM is required for Ca2+ mobilization and T cell proliferation. NSM-deficient T cells lack sustained CD3ζ and ZAP-70 phosphorylation and are unable to polarize and stabilize their microtubular system. We identified PKCζ as the key NSM downstream effector in this second wave of TCR signaling supporting dynamics of microtubule-organizing center (MTOC). Ceramide supplementation rescued PKCζ membrane recruitment and MTOC translocation in NSM-deficient cells. These findings identify the NSM as essential in TCR signaling when dynamic cytoskeletal reorganization promotes continued lateral and vertical supply of TCR signaling components: CD3ζ, Zap70, and PKCζ, and functional immune synapses are organized and stabilized via MTOC polarization.
Collapse
Affiliation(s)
- Charlene Börtlein
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Annette Draeger
- Department of Cell Biology, Institute for Anatomy, University of Bern, Bern, Switzerland
| | - Roman Schoenauer
- Department of Cell Biology, Institute for Anatomy, University of Bern, Bern, Switzerland
| | - Alexander Kuhlemann
- Department of Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | | | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
11
|
Guégan JP, Legembre P. Nonapoptotic functions of Fas/CD95 in the immune response. FEBS J 2017; 285:809-827. [PMID: 29032605 DOI: 10.1111/febs.14292] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/26/2017] [Accepted: 10/11/2017] [Indexed: 12/26/2022]
Abstract
CD95 (also known as Fas) is a member of the tumor necrosis factor receptor (TNFR) superfamily. Its cognate ligand, CD95L, is implicated in immune homeostasis and immune surveillance. Mutations in this receptor are associated with a loss of apoptotic signaling and have been detected in an autoimmune disorder called autoimmune lymphoproliferative syndrome (ALPS) type Ia, which shares some clinical features with systemic lupus erythematosus (SLE). In addition, deletions and mutations of CD95 have been described in many cancers, which led researchers to initially classify this receptor as a tumor suppressor. More recent data demonstrate that CD95 engagement evokes nonapoptotic signals that promote inflammation and carcinogenesis. Transmembrane CD95L (m-CD95L) can be cleaved by metalloproteases, releasing a soluble ligand (s-CD95L). Soluble and membrane-bound CD95L show different stoichiometry (homotrimer versus multimer of homotrimers, respectively), which differentially affects CD95-mediated signaling through molecular mechanisms that remain to be elucidated. This review discusses the biological roles of CD95 in light of recent experiments addressing how a death receptor can trigger both apoptotic and nonapoptotic signaling pathways.
Collapse
Affiliation(s)
- Jean-Philippe Guégan
- Centre Eugène Marquis, INSERM U1242-COSS, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France.,Université de Rennes-1, Rennes, France
| | - Patrick Legembre
- Centre Eugène Marquis, INSERM U1242-COSS, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France.,Université de Rennes-1, Rennes, France
| |
Collapse
|
12
|
Kamei R, Tanaka HY, Kawano T, Morii C, Tanaka S, Nishihara H, Iwata C, Kano MR. Regulation of endothelial Fas expression as a mechanism of promotion of vascular integrity by mural cells in tumors. Cancer Sci 2017; 108:1080-1088. [PMID: 28247971 PMCID: PMC5448593 DOI: 10.1111/cas.13216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/18/2017] [Accepted: 02/23/2017] [Indexed: 11/29/2022] Open
Abstract
Angiogenesis is a multi‐step process that culminates in vascular maturation whereby nascent vessels stabilize to become functional, and mural cells play an essential role in this process. Recent studies have shown that mural cells in tumors also promote and maintain vascular integrity, with wide‐reaching clinical implications including the regulation of tumor growth, metastases, and drug delivery. Various regulatory signaling pathways have been hitherto implicated, but whether regulation of Fas‐dependent apoptotic mechanisms is involved has not yet been fully investigated. We first compared endothelial FAS staining in human pancreatic ductal adenocarcinomas and colon carcinomas and show that the latter, characterized by lower mural cell coverage of tumor vasculature, demonstrated higher expression of FAS than the former. Next, in an in vitro coculture system of MS‐1 and 10T1/2 cells as endothelial and mural cells respectively, we show that mural cells decreased endothelial Fas expression. Then, in an in vivo model in which C26 colon carcinoma cells were inoculated together with MS‐1 cells alone or with the further addition of 10T1/2 cells, we demonstrate that mural cells prevented hemorrhage. Finally, knockdown of endothelial Fas sufficiently recapitulated the protection against hemorrhage seen with the addition of mural cells. These results together suggest that regulation of endothelial Fas signaling is involved in the promotion of vascular integrity by mural cells in tumors.
Collapse
Affiliation(s)
- Ryosuke Kamei
- Medical Scientist Training Program, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyoshi Y Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takao Kawano
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chiharu Morii
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Sayaka Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Nishihara
- Laboratory of Translational Pathology, Hokkaido University School of Medicine, Sapporo, Japan
| | - Caname Iwata
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsunobu R Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
13
|
Pharmacological targeting of ion channels for cancer therapy: In vivo evidences. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1385-97. [DOI: 10.1016/j.bbamcr.2015.11.032] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/29/2022]
|
14
|
Combs DJ, Lu Z. Sphingomyelinase D inhibits store-operated Ca2+ entry in T lymphocytes by suppressing ORAI current. ACTA ACUST UNITED AC 2016. [PMID: 26216860 PMCID: PMC4516786 DOI: 10.1085/jgp.201511359] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Sphingomyelinase D suppresses Orai current in human T cells and decreases cytokine production, providing a mechanism whereby certain bacteria could inhibit the immune system. Infections caused by certain bacteria including Mycobacterium tuberculosis and Corynebacterium pseudotuberculosis provoke inflammatory responses characterized by the formation of granulomas with necrotic foci—so-called caseous necrosis. The granulomas of infected animals show prominent infiltration by T lymphocytes, and T cell depletion increases host mortality. Notorious zoonotic C. pseudotuberculosis secretes sphingomyelinase (SMase) D, a phospholipase that cleaves off the choline moiety of sphingomyelin, a phospholipid found primarily in the outer leaflet of host cell plasma membranes. Experimental C. pseudotuberculosis strains that lack SMase D are markedly less infectious and unable to spread in hosts, indicating that this enzyme is a crucial virulence factor for sustaining the caseous lymphadenitis infections caused by this microbe. However, the molecular mechanism by which SMase D helps bacteria evade the host’s immune response remains unknown. Here, we find that SMase D inhibits store-operated Ca2+ entry (SOCE) in human T cells and lowers the production of the SOCE-dependent cytokines interleukin-2, which is critical for T cell growth, proliferation, and differentiation, and tumor necrosis factor α, which is crucial for the formation and maintenance of granulomas in microbial infections. SMase D inhibits SOCE through a previously unknown mechanism, namely, suppression of Orai1 current, rather than through altering gating of voltage-gated K+ channels. This finding suggests that, whereas certain genetic mutations abolish Orai1 activity causing severe combined immunodeficiency (SCID), bacteria have the ability to suppress Orai1 activity with SMase D to create an acquired, chronic SCID-like condition that allows persistent infection. Thus, in an example of how virulence factors can disrupt key membrane protein function by targeting phospholipids in host cell membranes, our study has uncovered a novel molecular mechanism that bacteria can use to thwart host immunity.
Collapse
Affiliation(s)
- David J Combs
- Department of Physiology, Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Zhe Lu
- Department of Physiology, Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
15
|
Collenburg L, Walter T, Burgert A, Müller N, Seibel J, Japtok L, Kleuser B, Sauer M, Schneider-Schaulies S. A Functionalized Sphingolipid Analogue for Studying Redistribution during Activation in Living T Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:3951-62. [DOI: 10.4049/jimmunol.1502447] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/02/2016] [Indexed: 11/19/2022]
|
16
|
Saslowsky DE, Thiagarajah JR, McCormick BA, Lee JC, Lencer WI. Microbial sphingomyelinase induces RhoA-mediated reorganization of the apical brush border membrane and is protective against invasion. Mol Biol Cell 2016; 27:1120-30. [PMID: 26864627 PMCID: PMC4814219 DOI: 10.1091/mbc.e15-05-0293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 02/01/2016] [Indexed: 12/19/2022] Open
Abstract
Both commensal and pathogenic microbes that colonize the GI tract can synthesize and secrete spingomyelinase enzymes that cleave membrane sphingomyelin, leaving the ceramide component intact in the cell membrane. This study examines how this reaction affects the structure and function of host enterocytes and mucosal defense. The apical brush border membrane (BBM) of intestinal epithelial cells forms a highly structured and dynamic environmental interface that serves to regulate cellular physiology and block invasion by intestinal microbes and their products. How the BBM dynamically responds to pathogenic and commensal bacterial signals can define intestinal homeostasis and immune function. We previously found that in model intestinal epithelium, the conversion of apical membrane sphingomyelin to ceramide by exogenous bacterial sphingomyelinase (SMase) protected against the endocytosis and toxicity of cholera toxin. Here we elucidate a mechanism of action by showing that SMase induces a dramatic, reversible, RhoA-dependent alteration of the apical cortical F-actin network. Accumulation of apical membrane ceramide is necessary and sufficient to induce the actin phenotype, and this coincides with altered membrane structure and augmented innate immune function as evidenced by resistance to invasion by Salmonella.
Collapse
Affiliation(s)
- David E Saslowsky
- Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA 02115 Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Harvard Medical School, Boston, MA 02115
| | - Jay R Thiagarajah
- Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA 02115 Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Harvard Medical School, Boston, MA 02115
| | - Beth A McCormick
- Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jean C Lee
- Harvard Medical School, Boston, MA 02115 Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115
| | - Wayne I Lencer
- Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA 02115 Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Harvard Medical School, Boston, MA 02115
| |
Collapse
|
17
|
Abstract
Acid sphingomyelinase (ASM), a lipid hydrolase enzyme, has the potential to modulate various cellular activation responses via the generation of ceramide and by interaction with cellular receptors. We have hypothesized that ASM modulates CD4+ T-cell receptor activation and impacts immune responses. We first observed interactions of ASM with the intracellular domains of both CD3 and CD28. ASM further mediates T-cell proliferation after anti-CD3/CD28 antibody stimulation and alters CD4+ T-cell activation signals by generating ceramide. We noted that various pharmacological inhibitors of ASM or knockdown of ASM using small hairpin RNA inhibit CD3/CD28-mediated CD4+ T-cell proliferation and activation. Furthermore, such blockade of ASM bioactivity by biochemical inhibitors and/or molecular-targeted knockdown of ASM broadly abrogate T-helper cell responses. In conclusion, we detail immune, pivotal roles of ASM in adaptive immune T-cell responses, and propose that these pathways might provide novel targets for the therapy of autoimmune and inflammatory diseases.
Collapse
|
18
|
Aureli M, Murdica V, Loberto N, Samarani M, Prinetti A, Bassi R, Sonnino S. Exploring the link between ceramide and ionizing radiation. Glycoconj J 2015; 31:449-59. [PMID: 25129488 DOI: 10.1007/s10719-014-9541-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The aim of radiotherapy is to eradicate cancer cells with ionizing radiation; tumor cell death following irradiation can be induced by several signaling pathways, most of which are triggered as a consequence of DNA damage, the primary and major relevant cell response to radiation. Several lines of evidence demonstrated that ceramide, a crucial sensor and/or effector of different signalling pathways promoting cell cycle arrest, death and differentiation, is directly involved in the molecular mechanisms underlying cellular response to irradiation. Most of the studies strongly support a direct relationship between ceramide accumulation and radiation-induced cell death, mainly apoptosis; for this reason, defining the contribution of the multiple metabolic pathways leading to ceramide formation and the causes of its dysregulated metabolism represent the main goal in order to elucidate the ceramide-mediated signaling in radiotherapy. In this review, we summarize the current knowledge concerning the different routes leading to ceramide accumulation in radiation-induced cell response with particular regard to the role of the enzymes involved in both ceramide neogenesis and catabolism. Emphasis is placed on sphingolipid breakdown as mechanism of ceramide generation activated following cell irradiation; the functional relevance of this pathway, and the role of glycosphingolipid glycohydrolases as direct targets of ionizing radiation are also discussed. These new findings add a further attractive point of investigation to better define the complex interplay between sphingolipid metabolism and radiation therapy.
Collapse
Affiliation(s)
- Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Via Fratelli Cervi 93, 20090, Segrate, Italy
| | | | | | | | | | | | | |
Collapse
|
19
|
Beyersdorf N, Müller N. Sphingomyelin breakdown in T cells: role in activation, effector functions and immunoregulation. Biol Chem 2015; 396:749-58. [DOI: 10.1515/hsz-2014-0282] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/03/2015] [Indexed: 11/15/2022]
Abstract
Abstract
Host T cell activation, a key step in obtaining adaptive immunity against pathogens, is initiated by the binding of the T cell receptor to a foreign antigenic peptide presented by the major histocompatibility complex on the surface of an antigen-presenting cell and, consequently, formation of an immunological synapse. Within the immunological synapse, the engagement of the T cell receptor in cooperation with simultaneous ligation of co-stimulatory molecules induces a precisely organized cascade of signaling events and pathways that regulate clonal expansion and differentiation of naïve T cells into effector T cells contributing to pathogen clearance. The biochemical changes that underlie T cell activation and differentiation, however, not only involve proteins but also lipids. In particular, catabolic cleavage of sphingomyelin generating ceramide can substantially influence functional responses in cells of the immune system. Changes in sphingomyelin and ceramide content have been reported to directly impact on membrane physiology, thus modifying signal transmission and interfering with diverse aspects of T cell activity. In this review we will focus on sphingomyelin breakdown/ceramide generation in T cells with regard to their function and development of T cell-mediated immunity.
Collapse
|
20
|
Gulbins E, Walter S, Becker KA, Halmer R, Liu Y, Reichel M, Edwards MJ, Müller CP, Fassbender K, Kornhuber J. A central role for the acid sphingomyelinase/ceramide system in neurogenesis and major depression. J Neurochem 2015; 134:183-92. [DOI: 10.1111/jnc.13145] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/15/2015] [Accepted: 04/09/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Erich Gulbins
- Department of Surgery; University of Cincinnati; Cincinnati Ohio USA
- Department of Molecular Biology; University of Duisburg-Essen; Essen Germany
| | - Silke Walter
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Katrin Anne Becker
- Department of Molecular Biology; University of Duisburg-Essen; Essen Germany
| | - Ramona Halmer
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Yang Liu
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy; Friedrich-Alexander-University of Erlangen; Erlangen Germany
| | | | - Christian P. Müller
- Department of Psychiatry and Psychotherapy; Friedrich-Alexander-University of Erlangen; Erlangen Germany
| | - Klaus Fassbender
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy; Friedrich-Alexander-University of Erlangen; Erlangen Germany
| |
Collapse
|
21
|
Managò A, Becker KA, Carpinteiro A, Wilker B, Soddemann M, Seitz AP, Edwards MJ, Grassmé H, Szabò I, Gulbins E. Pseudomonas aeruginosa pyocyanin induces neutrophil death via mitochondrial reactive oxygen species and mitochondrial acid sphingomyelinase. Antioxid Redox Signal 2015; 22:1097-110. [PMID: 25686490 PMCID: PMC4403017 DOI: 10.1089/ars.2014.5979] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Pulmonary infections with Pseudomonas aeruginosa are a serious clinical problem and are often lethal. Because many strains of P. aeruginosa are resistant to antibiotics, therapeutic options are limited. Neutrophils play an important role in the host's early acute defense against pulmonary P. aeruginosa. Therefore, it is important to define the mechanisms by which P. aeruginosa interacts with host cells, particularly neutrophils. RESULTS Here, we report that pyocyanin, a membrane-permeable pigment and toxin released by P. aeruginosa, induces the death of wild-type neutrophils; its interaction with the mitochondrial respiratory chain results in the release of reactive oxygen species (ROS), the activation of mitochondrial acid sphingomyelinase, the formation of mitochondrial ceramide, and the release of cytochrome c from mitochondria. A genetic deficiency in acid sphingomyelinase prevents both the activation of this pathway and pyocyanin-induced neutrophil death. This reduced death, on the other hand, is associated with an increase in the release of interleukin-8 from pyocyanin-activated acid sphingomyelinase-deficient neutrophils but not from wild-type cells. INNOVATION These studies identified the mechanisms by which pyocyanin induces the release of mitochondrial ROS and by which ROS induce neutrophil death via mitochondrial acid sphingomyelinase. CONCLUSION These findings demonstrate a novel mechanism of pyocyanin-induced death of neutrophils and show how this apoptosis balances innate immune reactions.
Collapse
|
22
|
Mueller N, Avota E, Collenburg L, Grassmé H, Schneider-Schaulies S. Neutral sphingomyelinase in physiological and measles virus induced T cell suppression. PLoS Pathog 2014; 10:e1004574. [PMID: 25521388 PMCID: PMC4270778 DOI: 10.1371/journal.ppat.1004574] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 11/12/2014] [Indexed: 11/20/2022] Open
Abstract
T cell paralysis is a main feature of measles virus (MV) induced immunosuppression. MV contact mediated activation of sphingomyelinases was found to contribute to MV interference with T cell actin reorganization. The role of these enzymes in MV-induced inhibition of T cell activation remained equally undefined as their general role in regulating immune synapse (IS) activity which relies on spatiotemporal membrane patterning. Our study for the first time reveals that transient activation of the neutral sphingomyelinase 2 (NSM2) occurs in physiological co-stimulation of primary T cells where ceramide accumulation is confined to the lamellum (where also NSM2 can be detected) and excluded from IS areas of high actin turnover. Genetic ablation of the enzyme is associated with T cell hyper-responsiveness as revealed by actin dynamics, tyrosine phosphorylation, Ca2+-mobilization and expansion indicating that NSM2 acts to suppress overshooting T cell responses. In line with its suppressive activity, exaggerated, prolonged NSM2 activation as occurring in co-stimulated T cells following MV exposure was associated with aberrant compartmentalization of ceramides, loss of spreading responses, interference with accumulation of tyrosine phosphorylated protein species and expansion. Altogether, this study for the first time reveals a role of NSM2 in physiological T cell stimulation which is dampening and can be abused by a virus, which promotes enhanced and prolonged NSM2 activation to cause pathological T cell suppression. Though the ability of measles virus (MV) to impair T cell activation has long been known, it is mechanistically not well understood. We have shown earlier that MV can contact dependently trigger activation of sphingomyelinases which is known to affect compartmentalization of membrane lipids and proteins. Because these are particularly important in the activity of the immune synapse (IS), we investigated whether MV-induced sphingomyelinase activity would interfere at that level with T cell activation. Our study for the first time revealed that the neutral sphingomyelinase 2 (NSM2) is transiently activated in primary T cells by co-stimulation through CD3 and CD28, and that this does occur to dampen early T cell responses. The virus appears to exploit this inhibitory activity of the enzyme to suppress T cell activation by promoting an enhanced and prolonged NSM2 activation. These findings do not only assign a hitherto novel role of the NSM2 in regulating T cell responses, but also reveal a novel strategy for viral T cell suppression.
Collapse
Affiliation(s)
- Nora Mueller
- University of Würzburg, Institute for Virology and Immunobiology, Wuerzburg, Germany
| | - Elita Avota
- University of Würzburg, Institute for Virology and Immunobiology, Wuerzburg, Germany
| | - Lena Collenburg
- University of Würzburg, Institute for Virology and Immunobiology, Wuerzburg, Germany
| | | | | |
Collapse
|
23
|
Becker KA, Henry B, Ziobro R, Riethmüller J, Gulbins E. Lipids in cystic fibrosis. Expert Rev Respir Med 2014; 5:527-35. [DOI: 10.1586/ers.11.36] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
24
|
Abstract
Cell shrinkage is a hallmark and contributes to signaling of apoptosis. Apoptotic cell shrinkage requires ion transport across the cell membrane involving K(+) channels, Cl(-) or anion channels, Na(+)/H(+) exchange, Na(+),K(+),Cl(-) cotransport, and Na(+)/K(+)ATPase. Activation of K(+) channels fosters K(+) exit with decrease of cytosolic K(+) concentration, activation of anion channels triggers exit of Cl(-), organic osmolytes, and HCO3(-). Cellular loss of K(+) and organic osmolytes as well as cytosolic acidification favor apoptosis. Ca(2+) entry through Ca(2+)-permeable cation channels may result in apoptosis by affecting mitochondrial integrity, stimulating proteinases, inducing cell shrinkage due to activation of Ca(2+)-sensitive K(+) channels, and triggering cell-membrane scrambling. Signaling involved in the modification of cell-volume regulatory ion transport during apoptosis include mitogen-activated kinases p38, JNK, ERK1/2, MEKK1, MKK4, the small G proteins Cdc42, and/or Rac and the transcription factor p53. Osmosensing involves integrin receptors, focal adhesion kinases, and tyrosine kinase receptors. Hyperosmotic shock leads to vesicular acidification followed by activation of acid sphingomyelinase, ceramide formation, release of reactive oxygen species, activation of the tyrosine kinase Yes with subsequent stimulation of CD95 trafficking to the cell membrane. Apoptosis is counteracted by mechanisms involved in regulatory volume increase (RVI), by organic osmolytes, by focal adhesion kinase, and by heat-shock proteins. Clearly, our knowledge on the interplay between cell-volume regulatory mechanisms and suicidal cell death is still far from complete and substantial additional experimental effort is needed to elucidate the role of cell-volume regulatory mechanisms in suicidal cell death.
Collapse
Affiliation(s)
- Florian Lang
- Institute of Physiology, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
25
|
Corre I, Guillonneau M, Paris F. Membrane signaling induced by high doses of ionizing radiation in the endothelial compartment. Relevance in radiation toxicity. Int J Mol Sci 2013; 14:22678-96. [PMID: 24252908 PMCID: PMC3856084 DOI: 10.3390/ijms141122678] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/01/2013] [Accepted: 11/06/2013] [Indexed: 01/30/2023] Open
Abstract
Tumor areas can now be very precisely delimited thanks to technical progress in imaging and ballistics. This has also led to the development of novel radiotherapy protocols, delivering higher doses of ionizing radiation directly to cancer cells. Despite this, radiation toxicity in healthy tissue remains a major issue, particularly with dose-escalation in these new protocols. Acute and late tissue damage following irradiation have both been linked to the endothelium irrigating normal tissues. The molecular mechanisms involved in the endothelial response to high doses of radiation are associated with signaling from the plasma membrane, mainly via the acid sphingomyelinase/ceramide pathway. This review describes this signaling pathway and discusses the relevance of targeting endothelial signaling to protect healthy tissues from the deleterious effects of high doses of radiation.
Collapse
Affiliation(s)
- Isabelle Corre
- CRCNA-UMR Inserm U892-CNRS 6299-Institut de Recherche en Santé de l'Université de Nantes, Nantes 44007, France.
| | | | | |
Collapse
|
26
|
Brint E, O’Callaghan G, Houston A. Life in the Fas lane: differential outcomes of Fas signaling. Cell Mol Life Sci 2013; 70:4085-99. [PMID: 23579628 PMCID: PMC11113183 DOI: 10.1007/s00018-013-1327-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 03/14/2013] [Accepted: 03/18/2013] [Indexed: 12/11/2022]
Abstract
Fas, also known as CD95 or APO-1, is a member of the tumor necrosis factor/nerve growth factor superfamily. Although best characterized in terms of its apoptotic function, recent studies have identified several other cellular responses emanating from Fas. These responses include migration, invasion, inflammation, and proliferation. In this review, we focus on the diverse cellular outcomes of Fas signaling and the molecular switches identified to date that regulate its pro- and anti-apoptotic functions. Such switches occur at different levels of signal transduction, ranging from the receptor through to cross-talk with other signaling pathways. Factors identified to date including other extracellular signals, proteins recruited to the death-inducing signaling complex, and the availability of different intracellular components of signal transduction pathways. The success of therapeutically targeting Fas will require a better understanding of these pathways, as well as the regulatory mechanisms that determine cellular outcome following receptor activation.
Collapse
Affiliation(s)
- Elizabeth Brint
- Department of Pathology, University College Cork, National University of Ireland, Cork, Ireland
| | - Grace O’Callaghan
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| |
Collapse
|
27
|
Kilgore JA, Dolman NJ, Davidson MW. A review of reagents for fluorescence microscopy of cellular compartments and structures, Part II: reagents for non-vesicular organelles. ACTA ACUST UNITED AC 2013; 66:12.31.1-12.31.24. [PMID: 24510724 DOI: 10.1002/0471142956.cy1231s66] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A wide range of fluorescent dyes and reagents exist for labeling organelles in live and fixed cells. Choosing between them can sometimes be confusing, and optimization for many of them can be challenging. Presented here is a discussion on the commercially-available reagents that have shown the most promise for each organelle of interest, including endoplasmic reticulum/nuclear membrane, Golgi apparatus, mitochondria, nucleoli, and nuclei, with an emphasis on localization of these structures for microscopy. Included is a featured reagent for each structure with a recommended protocol, troubleshooting guide, and example image.
Collapse
Affiliation(s)
- Jason A Kilgore
- Molecular Probes Labeling and Detection, Life Technologies, Eugene, Oregon
| | - Nick J Dolman
- Molecular Probes Labeling and Detection, Life Technologies, Eugene, Oregon
| | - Michael W Davidson
- National High Magnetic Field Laboratory and Department of Biological Science, Florida State University, Tallahassee, Florida
| |
Collapse
|
28
|
Gulbins E, Palmada M, Reichel M, Lüth A, Böhmer C, Amato D, Müller CP, Tischbirek CH, Groemer TW, Tabatabai G, Becker KA, Tripal P, Staedtler S, Ackermann TF, van Brederode J, Alzheimer C, Weller M, Lang UE, Kleuser B, Grassmé H, Kornhuber J. Acid sphingomyelinase-ceramide system mediates effects of antidepressant drugs. Nat Med 2013; 19:934-8. [PMID: 23770692 DOI: 10.1038/nm.3214] [Citation(s) in RCA: 273] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 04/23/2013] [Indexed: 01/18/2023]
Abstract
Major depression is a highly prevalent severe mood disorder that is treated with antidepressants. The molecular targets of antidepressants require definition. We investigated the role of the acid sphingomyelinase (Asm)-ceramide system as a target for antidepressants. Therapeutic concentrations of the antidepressants amitriptyline and fluoxetine reduced Asm activity and ceramide concentrations in the hippocampus, increased neuronal proliferation, maturation and survival and improved behavior in mouse models of stress-induced depression. Genetic Asm deficiency abrogated these effects. Mice overexpressing Asm, heterozygous for acid ceramidase, treated with blockers of ceramide metabolism or directly injected with C16 ceramide in the hippocampus had higher ceramide concentrations and lower rates of neuronal proliferation, maturation and survival compared with controls and showed depression-like behavior even in the absence of stress. The decrease of ceramide abundance achieved by antidepressant-mediated inhibition of Asm normalized these effects. Lowering ceramide abundance may thus be a central goal for the future development of antidepressants.
Collapse
Affiliation(s)
- Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Azzam R, Hariri F, El-Hachem N, Kamar A, Dbaibo G, Nemer G, Bitar F. Regulation of de novo ceramide synthesis: the role of dihydroceramide desaturase and transcriptional factors NFATC and Hand2 in the hypoxic mouse heart. DNA Cell Biol 2013; 32:310-9. [PMID: 23672204 DOI: 10.1089/dna.2013.1993] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We have previously shown that ceramide, a proapoptotic molecule decreases in the mouse heart as it adapts to hypoxia. We have also shown that its precursor, dihydroceramide, accumulates with hypoxia. This implicates the enzyme dihydroceramide desaturase (DHC-DS), which converts dihydroceramide to ceramide, in a potential regulatory checkpoint in cardiomyocytes. We hypothesised that the regulation of de novo ceramide synthesis plays an important role in the cardiomyocyte adaptation to hypoxia. We used an established mouse model to induce acute and chronic hypoxia. Cardiac tissues were extracted and quantitative real-time polymerase chain reaction (qRT-PCR) was used to evaluate the expression levels of DHC-DS. Electrophoretic Mobility Shift Assays (EMSAs) and qRT-PCR were used to evaluate the activity and expression levels of an array of transcription factors that might regulate DEGS1 gene expression. We demonstrated that DEGS1 mRNA levels decrease with time in hypoxic mice concurrent with the decrease in HAND2 transcripts. Interestingly, the DEGS1 promoter harbors overlapping sites for Hand2 and Nuclear Factor of Activated T-cells (NFATC) transcription factors. We have demonstrated a physical interaction between NFATC1 and the E-Box proteins with EMSA and coimmunoprecipitation assays. The regulation of de novo ceramide synthesis in response to hypoxia and this newly described interaction between E-box and NFATC transcription factors will pave the way to identify new pathways in the adaptation of the cardiomyocyte to stress. The elucidation of these pathways will in the long-term provide insights into potential targets for novel therapeutic regimens.
Collapse
Affiliation(s)
- Raed Azzam
- Department of Pediatrics, American University of Beirut-Medical Center, Beirut, Lebanon
| | | | | | | | | | | | | |
Collapse
|
30
|
Sukumaran P, Lönnfors M, Långvik O, Pulli I, Törnquist K, Slotte JP. Complexation of c6-ceramide with cholesteryl phosphocholine - a potent solvent-free ceramide delivery formulation for cells in culture. PLoS One 2013; 8:e61290. [PMID: 23620740 PMCID: PMC3631171 DOI: 10.1371/journal.pone.0061290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/06/2013] [Indexed: 12/15/2022] Open
Abstract
Ceramides are potent bioactive molecules in cells. However, they are very hydrophobic molecules, and difficult to deliver efficiently to cells. We have made fluid bilayers from a short-chain D-erythro-ceramide (C6-Cer) and cholesteryl phosphocholine (CholPC), and have used this as a formulation to deliver ceramide to cells. C6-Cer complexed with CholPC led to much larger biological effects in cultured cells (rat thyroid FRTL-5 and human HeLa cells in culture) compared to C6-Cer dissolved in dimethyl sulfoxide (DMSO). Inhibition of cell proliferation and induction of apoptosis was significantly more efficient by C6-Cer/CholPC compared to C6-Cer dissolved in DMSO. C6-Cer/CholPC also permeated cell membranes and caused mitochondrial Ca2+ influx more efficiently than C6-Cer in DMSO. Even though CholPC was taken up by cells to some extent (from C6-Cer/CholPC bilayers), and was partially hydrolyzed to free cholesterol (about 9%), none of the antiproliferative effects were due to CholPC or excess cholesterol. The ceramide effect was not limited to D-erythro-C6-Cer, since L-erythro-C6-Cer and D-erythro-C6-dihydroCer also inhibited cell priolifereation and affected Ca2+ homeostasis. We conclude that C6-Cer complexed to CholPC increased the bioavailability of the short-chain ceramide for cells, and potentiated its effects in comparison to solvent-dissolved C6-Cer. This new ceramide formulation appears to be superior to previous solvent delivery approaches, and may even be useful with longer-chain ceramides.
Collapse
Affiliation(s)
| | - Max Lönnfors
- Biochemistry, Department of Biosciences, Åbo Akademi University, Turku, Finland
| | - Otto Långvik
- Laboratory of Organic Chemistry, Department of Natural Sciences, Åbo Akademi University, Turku, Finland
| | - Ilari Pulli
- Cell Biology, Åbo Akademi University, Turku, Finland
| | - Kid Törnquist
- Cell Biology, Åbo Akademi University, Turku, Finland
- Minerva Foundation Institute of Medical Research, Biomedicum Helsinki, Helsinki, Finland
- * E-mail: (KT); (JPS)
| | - J. Peter Slotte
- Biochemistry, Department of Biosciences, Åbo Akademi University, Turku, Finland
- * E-mail: (KT); (JPS)
| |
Collapse
|
31
|
Abstract
The enzyme acid sphingomyelinase catalyzes the hydrolysis of sphingomyelin to ceramide. The importance of the enzyme for cell functions was first recognized in Niemann-Pick disease type A and B, the genetic disorders with a massive accumulation of sphingomyelin in many organs. Studies in the last years demonstrated that the enzyme also has an important role in cell signalling. Thus, the acid sphingomyelinase has a central function for the re-organization of molecules within the cell upon stimulation and thereby for the response of cells to stress and the induction of cell death but also proliferation and differentiation. Here, we discuss the current state of the art of the structure, regulation, and function of the acid sphingomyelinase.
Collapse
Affiliation(s)
- Brian Henry
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | | | | | |
Collapse
|
32
|
Abstract
Besides their essential role in the immune system, sphingolipids and their metabolites are potential key regulators in the life cycle of obligatory intracellular pathogens such as viruses. They are involved in lateral and vertical segregation of receptors required for attachment, membrane fusion and endocytosis, as well as in the intracellular replication, assembly and release of viruses. Glycosphingolipids may themselves act as receptors for viruses, such as Galactosylceramide for human immunodeficiency virus (HIV). In addition, sphingolipids and their metabolites are inseparably interwoven in signal transduction processes, dynamic alterations of the cytoskeleton, and the regulation of innate and intrinsic responses of infected target cells. Depending on the nature of the intracellular pathogen, they may support or inhibit infections. Understanding of the underlying mechanisms depending on the specific virus, immune control, and type of disease may open new avenues for therapeutic interventions.
Collapse
|
33
|
Thangaraju S, Subramani E, Chakravarty B, Chaudhury K. Therapeutic targeting of the TNF superfamily: A promising treatment for advanced endometrial adenocarcinoma. Gynecol Oncol 2012; 127:426-32. [DOI: 10.1016/j.ygyno.2012.07.125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/26/2012] [Accepted: 07/29/2012] [Indexed: 12/18/2022]
|
34
|
Li X, Gulbins E, Zhang Y. Oxidative stress triggers Ca-dependent lysosome trafficking and activation of acid sphingomyelinase. Cell Physiol Biochem 2012; 30:815-26. [PMID: 22890197 PMCID: PMC3777434 DOI: 10.1159/000341460] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2012] [Indexed: 12/27/2022] Open
Abstract
Recent studies demonstrate that rapid translocation of the acid sphingomyelinase (ASM), a lysosomal hydrolase, to the outer leaflet of the cell membrane and concomitant release of ceramide constitute a common cellular signaling cascade to various stimuli including CD95 ligation, UV-irradiation, bacterial and viral infections. Reactive oxygen species (ROS) were shown to play a crucial role in regulating this signaling cascade at least for some bacterial infections and UV-irradiation. However, the precise role of ROS for regulation of ASM is unknown. Here, by confocal microscopy and flow cytometry analysis, we demonstrate that hydrogen peroxide (H(2)O(2)), a primary form of ROS in mammalian cells, induces very rapid translocation of ASM and formation of ceramide-enriched membrane platforms in the plasma membrane of Jurkat T cells. In parallel, H(2)O(2) triggers lysosome trafficking and fusion with the plasma membrane, i.e. lysosome exocytosis, as detected by exposure of a lysosome-associated protein, LAMP1. Depletion of intracellular Ca(2+) by cell permeable EGTA-AM inhibits H(2)O(2)-induced lysosome exocytosis, ASM translocation and formation of ceramide-enriched platforms. Pharmacological inhibition or genetic deficiency of ASM did not affect H(2)O(2)-induced lysosome exocytosis. These results indicate that ROS-induced membrane translocation of ASM is mediated by exocytosis of lysosomes, which is dependent on intracellular Ca(2+) release.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, VA 23298, U.S.A
| | - Erich Gulbins
- Institute of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, Essen, Germany
| | - Yang Zhang
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, VA 23298, U.S.A
| |
Collapse
|
35
|
Roy A, Sil PC. Taurine protects murine hepatocytes against oxidative stress-induced apoptosis by tert-butyl hydroperoxide via PI3K/Akt and mitochondrial-dependent pathways. Food Chem 2012. [DOI: 10.1016/j.foodchem.2011.09.057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
36
|
Differential effects of alprazolam and clonazepam on the immune system and blood vessels of non-stressed and stressed adult male albino rats. Interdiscip Toxicol 2011; 4:132-43. [PMID: 22058654 PMCID: PMC3203915 DOI: 10.2478/v10102-011-0021-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 07/01/2011] [Accepted: 07/21/2011] [Indexed: 01/21/2023] Open
Abstract
Benzodiazepines belongs to one of the most commonly used anxiolytic and anticonvulsant drugs in the world. Full description of toxic effects on different organs is lacking for nearly all the current benzodiazepines. The aim of the current work was to study the immunologic and vascular changes induced by sub-chronic administration of alprazolam and clonazepam in non-stressed and stressed adult male albino rats. Forty-two adult male albino rats were divided into 6 groups (I): (Ia) Negative control rats, (Ib): Positive control rats received distilled water, (II): Stressed rats, (III): Non-stressed rats received daily oral dose of clonazepam (0.5 mg/kg), (IV): Stressed rats received daily oral dose of clonazepam (0.5 mg/kg), (V): Non-stressed rats received daily oral dose of alprazolam (0.3 mg/kg). (VI): Stressed rats received daily oral dose of alprazolam (0.3 mg/kg). At the end of the 4th week, total leukocyte count (WBCs) and differential count were determined, anti-sheep RBC antibody (Anti-SRBC) titer and interleukin-2 (IL-2) level were assessed, thymus glands, lymph nodes, spleens and abdominal aortae were submitted to histopathological examination. Alprazolam was found to induce a significant increase in neutrophil count and a significant decrease in lymphocytes, anti-SRBC titer and IL-2 level with severe depletion of the splenic, thymal and nodal lymphocytes, accompanied by congestion and eosinophilic vasculitis of all organs tested in comparison to clonazepam treated rats. Stress enhanced the toxic effects. It was concluded that the immune system and blood vessels can be adversely affected to a greater extent by short-term chronic administration of alprazolam than by clonazepam, and these toxic effects are aggravated by stress.
Collapse
|
37
|
ORAI-mediated calcium influx in T cell proliferation, apoptosis and tolerance. Cell Calcium 2011; 50:261-9. [DOI: 10.1016/j.ceca.2011.05.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 12/25/2022]
|
38
|
Abstract
Death receptors play a crucial role in immune surveillance and cellular homeostasis, two processes circumvented by tumor cells. CD95 (also termed Fas or APO1) is a transmembrane receptor, which belongs to the tumor necrosis factor receptor superfamily, and induces a potent apoptotic signal. Initial steps of the CD95 signal take place through protein/protein interactions that bring zymogens such as caspase-8 and caspase-10 closer. Aggregation of these procaspases leads to their autoprocessing, to the release of activated caspases in the cytosol, which causes a caspase cascade, and to the transmission of the apoptotic signal. In parallel, CD95 engagement drives an increase in the intracellular calcium concentration (Ca(2+))i whose origin and functions remain controversial. Although Ca(2+) ions play a central role in apoptosis/necrosis induction, recent studies have highlighted a protective role of Ca(2+) in death receptor signaling. In the light of these findings, we discuss the role of Ca(2+) ions as modulators of CD95 signaling.
Collapse
|
39
|
Henry B, Möller C, Dimanche-Boitrel MT, Gulbins E, Becker KA. Targeting the ceramide system in cancer. Cancer Lett 2011; 332:286-94. [PMID: 21862212 DOI: 10.1016/j.canlet.2011.07.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 06/30/2011] [Accepted: 07/08/2011] [Indexed: 12/20/2022]
Abstract
Sphingolipids, in particular ceramide, have been described as important components of cellular signalling pathways. Ceramide can be produced via multiple mechanisms including through the hydrolysis of sphingomyelin by acid and neutral sphingomyelinase or by a de novo synthesis pathway. Recent studies have identified sphingomyelinases and ceramide synthases as important targets for γ-irradiation and chemotherapeutic drugs. Likewise, common cancer treatment modalities, such as γ-irradiation and many chemotherapeutic agents, induce cell death via the generation of ceramide. This suggests that the manipulation of ceramide production and metabolism could offer promising means for the enhancement of anti-tumor therapies. The focus of this mini-review will be to discuss contemporary evidence suggesting that ceramide forming pathways and ceramide itself are important targets for the treatment of tumors and the development of novel tumor treatment strategies.
Collapse
Affiliation(s)
- Brian Henry
- Dept. of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45 122 Essen, Germany
| | | | | | | | | |
Collapse
|
40
|
Lang F, Ullrich S, Gulbins E. Ceramide formation as a target in beta-cell survival and function. Expert Opin Ther Targets 2011; 15:1061-71. [PMID: 21635197 DOI: 10.1517/14728222.2011.588209] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Ceramide may be synthesized de novo or generated by sphingomyelinase-dependent hydrolysis of sphingomyelin. AREAS COVERED The role of ceramide, ceramide-sensitive signaling and ion channels in β-cell apoptosis, lipotoxicity and amyloid-induced β-cell death. EXPERT OPINION Ceramide participates in β-cell dysfunction and apoptosis after exposure to TNFα, IL-1β and IFN-γ, excessive amyloid and islet amyloid polypeptide or non-esterified fatty acids (lipotoxicity). Knockout of sphingomyelin synthase 1, which converts ceramide to sphingomyelin, leads to impairment of insulin secretion. Increased ceramidase activity or pharmacological inhibition of ceramide synthetase, inhibits β-cell apoptosis. Ceramide contributes to endoplasmatic reticulum (ER) stress, decreased mitochondrial membrane potential in insulin-secreting cells and mitochondrial release of cytochrome c into the cytosol, which are all triggers of apoptotic cell death. Ceramide-dependent signaling involves activation of extracellularly regulated kinases 1 and 2 (ERK1/2), downregulation of Period (Per)-aryl hydrocarbon receptor nuclear translocator (Arnt)-single-minded (Sim) kinase (PASK), activation of okadaic-acid-sensitive protein phosphatase 2A (PP2A) and stimulation of NADPH-oxidase with generation of superoxides and lipid peroxides. Ceramide reduces the activity of voltage gated potassium (Kv)-channels in insulin-secreting cells. The role of ceramide in β-cell survival and function may be therapeutically relevant, because ceramide formation can be suppressed by pharmacological inhibition of ceramide synthetase and/or sphingomyelinase.
Collapse
Affiliation(s)
- Florian Lang
- University of Tübingen, Institute of Physiology, Germany.
| | | | | |
Collapse
|
41
|
Onopiuk M, Wierzbicka K, Brutkowski W, Szczepanowska J, Zabłocki K. Caspase-dependent inhibition of store-operated Ca(2+) entry into apoptosis-committed Jurkat cells. Biochem Biophys Res Commun 2010; 399:198-202. [PMID: 20643097 DOI: 10.1016/j.bbrc.2010.07.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 07/15/2010] [Indexed: 11/15/2022]
Abstract
Activation of T-cells triggers store-operated Ca(2+) entry, which begins a signaling cascade leading to induction of appropriate gene expression and eventually lymphocyte proliferation and differentiation. The simultaneous enhancement of Fas ligand gene expression in activated cells allows the immune response to be limited by committing the activated cells to apoptosis. In apoptotic cells the store-operated calcium entry is significantly inhibited. It has been documented that moderate activation of Fas receptor may cause reversible inhibition of store-operated channels by ceramide released from hydrolyzed sphingomyelin. Here we show that activation of Fas receptor in T-cells results in caspase-dependent decrease of cellular STIM1 and Orai1 protein content. This effect may be responsible for the substantial inhibition of Ca(2+) entry into Jurkat cells undergoing apoptosis. In turn, this inhibition might prevent overloading of cells with calcium and protect them against necrosis.
Collapse
Affiliation(s)
- Marta Onopiuk
- Department of Biochemistry, The Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | | | | | |
Collapse
|
42
|
The role of FasL and Fas in health and disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 647:64-93. [PMID: 19760067 DOI: 10.1007/978-0-387-89520-8_5] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The FS7-associated cell surface antigen (Fas, also named CD95, APO-1 or TNFRSF6) attracted considerable interest in the field of apoptosis research since its discovery in 1989. The groups of Shin Yonehara and Peter Krammer were the first reporting extensive apoptotic cell death induction upon treating cells with Fas-specific monoclonal antibodies.1,2 Cloning of Fas3 and its ligand,4,5 FasL (also known as CD178, CD95L or TNFSF6), laid the cornerstone in establishing this receptor-ligand system as a central regulator of apoptosis in mammals. Therapeutic exploitation of FasL-Fas-mediated cytotoxicity was soon an ambitous goal and during the last decade numerous strategies have been developed for its realization. In this chapter, we will briefly introduce essential general aspects of the FasL-Fas system before reviewing its physiological and pathophysiological relevance. Finally, FasL-Fas-related therapeutic tools and concepts will be addressed.
Collapse
|
43
|
Role of Kv1.3 mitochondrial potassium channel in apoptotic signalling in lymphocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1251-9. [DOI: 10.1016/j.bbabio.2010.01.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 01/14/2010] [Accepted: 01/14/2010] [Indexed: 11/24/2022]
|
44
|
Becker KA, Riethmüller J, Zhang Y, Gulbins E. The role of sphingolipids and ceramide in pulmonary inflammation in cystic fibrosis. Open Respir Med J 2010; 4:39-47. [PMID: 20556203 DOI: 10.2174/1874306401004020039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 10/20/2009] [Accepted: 10/21/2009] [Indexed: 01/01/2023] Open
Abstract
Sphingolipids and in particular ceramide have been shown to be critically involved in the response to many receptor-mediated, but also receptor-independent, mainly stress stimuli. Recent studies demonstrate that ceramide plays an important role in the pathogenesis of cystic fibrosis, a hereditary metabolic disorder caused by mutations of the Cystic Fibrosis Transmembrane Conductance Regulator. Patients with cystic fibrosis suffer from chronic pulmonary inflammation and microbial lung infections, in particular with Pseudomonas aeruginosa. Chronic pulmonary inflammation in these patients seems to be the initial pathophysiological event. Inflammation may finally result in the high infection susceptibility of these patients, fibrosis and loss of lung function. Recent studies demonstrated that ceramide accumulates in lungs of cystic fibrosis mice and causes age-dependent pulmonary inflammation as indicated by accumulation of neutrophils and macrophages in the lung and increased pulmonary concentrations of Interleukins 1 and 8, death of bronchial epithelial cells, deposition of DNA in bronchi and high susceptibility to Pseudomonas aeruginosa infections. Genetic or pharmacological inhibition of the acid sphingomyelinase blocks excessive ceramide production in lungs of cystic fibrosis mice and corrects pathological lung findings. First clinical studies confirm that inhibition of the acid sphingomyelinase with small molecules might be a novel strategy to treat patients with cystic fibrosis.
Collapse
Affiliation(s)
- Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| | | | | | | |
Collapse
|
45
|
Zhang Y, Li X, Grassmé H, Döring G, Gulbins E. Alterations in ceramide concentration and pH determine the release of reactive oxygen species by Cftr-deficient macrophages on infection. THE JOURNAL OF IMMUNOLOGY 2010; 184:5104-11. [PMID: 20351190 DOI: 10.4049/jimmunol.0902851] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We recently demonstrated that the accumulation of ceramide in Cftr-deficient epithelial cells is important for the pathophysiology of CF. However, the role of ceramide in other lung cells, particularly lung macrophages, requires definition. In this study, we report that ceramide is accumulated in Cftr-deficient lung macrophages. Alveolar macrophages contain a vesicle population, which is stained with LysoSensor probes but not by tetramethylrhodamine dextran. These vesicles, presumably secretory lysosomes, exhibit a higher pH in Cftr-deficient macrophages than the corresponding vesicles in lung macrophages isolated from wild-type (WT) mice. Alkalinization of these vesicles in Cftr-deficient macrophages correlates with a failure of the macrophages to respond to infection with various Pseudomonas aeruginosa strains by acutely activating acid sphingomyelinase, releasing ceramide, forming ceramide-enriched membrane platforms that serve to cluster gp91(phox), and, most importantly, releasing reactive oxygen species (ROS). In contrast, these events occur rapidly in WT lung macrophages postinfection. Inhibiting ROS in WT macrophages prevents the killing of P. aeruginosa. These findings provide evidence for a novel pH-controlled pathway from acid sphingomyelinase activation via ceramide and clustering of gp91(phox) to the release of ROS in lung macrophages.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | | | | | | | | |
Collapse
|
46
|
Gatidis S, Borst O, Föller M, Lang F. Effect of osmotic shock and urea on phosphatidylserine scrambling in thrombocyte cell membranes. Am J Physiol Cell Physiol 2010; 299:C111-8. [PMID: 20237147 DOI: 10.1152/ajpcell.00477.2009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Blood passing the renal medulla enters a strongly hypertonic environment challenging functional properties and survival of blood cells. In erythrocytes, exposure to hyperosmotic shock stimulates Ca(2+) entry and ceramide formation with subsequent cell membrane scrambling, an effect partially reversed by high concentrations of Cl(-) or urea. Cell membrane scrambling with phosphatidylserine exposure is part of the procoagulant phenotype of platelets. Coagulation in the hypertonic renal medulla would jeopardize blood flow in the vasa recta. The present study thus explored whether hypertonic environment and urea modify phosphatidylserine exposure of human platelets. FACS analysis was employed to estimate cytosolic Ca(2+) activity with Fluo3 fluorescence, ceramide formation, P-selectin, and glycoprotein IIb/IIIa activation with fluorescent antibodies and phosphatidylserine exposure with annexin V-binding. The spontaneous platelet aggregation was measured by impedance aggregometry. Hyperosmotic shock (addition of 500 mM sucrose or 250 mM NaCl) significantly enhanced cytosolic Ca(2+) activity, ceramide formation, phosphatidylserine exposure, platelet degranulation, and aggregability. Addition of 500 mM urea to isotonic saline did not significantly modify cytosolic Ca(2+) activity, ceramide abundance, or annexin V-binding but significantly blunted the respective effects of hypertonic shock following addition of 500 mM sucrose. In isotonic solutions, both ceramide (20 microM) and Ca(2+) ionophore ionomycin (0.5 microM) increased annexin V-binding, effects again significantly blunted by 500 mM urea. Moreover, oxidative stress by addition of 0.5 mM peroxynitrite increased cytosolic Ca(2+) activity and triggered annexin V-binding, effects again blunted in the presence of 500 mM urea. The observations reveal that hyperosmotic shock and oxidative stress trigger a procoagulant platelet phenotype, an effect blunted by the presence of high urea concentrations.
Collapse
Affiliation(s)
- Sergios Gatidis
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | |
Collapse
|
47
|
Kornhuber J, Reichel M, Tripal P, Groemer TW, Henkel AW, Mühle C, Gulbins E. The role of ceramide in major depressive disorder. Eur Arch Psychiatry Clin Neurosci 2009; 259 Suppl 2:S199-204. [PMID: 19876679 DOI: 10.1007/s00406-009-0061-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Major depression is a severe mood disorder with a lifetime prevalence of more than 10%. The pharmacokinetic hypothesis claims that a slow accumulation of antidepressant drugs by acid trapping mainly into lysosomes is responsible for the therapeutic latency and that a lysosomal target mediates the antidepressant effects. The lysosomal lipid metabolizing enzyme acid sphingomyelinase (ASM) cleaves sphingomyelin into ceramide and phosphorylcholine. In a pilot study, the activity of this enzyme was increased in peripheral blood cells of patients with major depressive disorder (MDD), making the ASM an interesting molecular target of antidepressant drugs. Indeed, several antidepressant drugs functionally inhibit ASM. The ASM/ceramide pathway might be a missing link unifying independent findings in neurobiology and the treatment of MDD such as therapeutic latency, oxidative stress, immune activation and increased risk of cardiovascular disease.
Collapse
Affiliation(s)
- Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University of Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany.
| | | | | | | | | | | | | |
Collapse
|
48
|
Valencia-Cruz G, Shabala L, Delgado-Enciso I, Shabala S, Bonales-Alatorre E, Pottosin II, Dobrovinskaya OR. K(bg) and Kv1.3 channels mediate potassium efflux in the early phase of apoptosis in Jurkat T lymphocytes. Am J Physiol Cell Physiol 2009; 297:C1544-53. [PMID: 19794143 DOI: 10.1152/ajpcell.00064.2009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Microelectrode ion flux estimation (MIFE) and patch-clamp techniques were combined for noninvasive K(+) flux measurements and recording of activities of the dominant K(+) channels in the early phases of apoptosis in Jurkat cells. Staurosporine (STS, 1 microM) evoked rapid (peaking around 15 min) transient K(+) efflux, which then gradually decreased. This transient K(+) efflux occurred concurrently with the transient increase of the K(+) background (K(bg)) TWIK-related spinal cord K(+) channel-like current density, followed by a drastic decrease and concomitant membrane depolarization. The Kv1.3 current density remained almost constant. Kv1.3 activation was not altered by STS, whereas the inactivation was shifted to more positive potentials. Contribution of K(bg) and Kv1.3 channels to the transient and posttransient STS-induced K(+) efflux components, respectively, was confirmed by the effects of bupivacaine, predominantly blocking K(bg) current, and the Kv1.3-specific blocker margatoxin. Channel-mediated K(+) efflux provoked a substantial cellular shrinkage and affected the activation of caspases.
Collapse
|
49
|
Sphingomyelinase dependent apoptosis following treatment of pancreatic beta-cells with amyloid peptides Aß1-42 or IAPP. Apoptosis 2009; 14:878-89. [DOI: 10.1007/s10495-009-0364-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
50
|
Strauss G, Lindquist JA, Arhel N, Felder E, Karl S, Haas TL, Fulda S, Walczak H, Kirchhoff F, Debatin KM. CD95 co-stimulation blocks activation of naive T cells by inhibiting T cell receptor signaling. ACTA ACUST UNITED AC 2009; 206:1379-93. [PMID: 19487421 PMCID: PMC2715064 DOI: 10.1084/jem.20082363] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CD95 is a multifunctional receptor that induces cell death or proliferation depending on the signal, cell type, and cellular context. Here, we describe a thus far unknown function of CD95 as a silencer of T cell activation. Naive human T cells triggered by antigen-presenting cells expressing a membrane-bound form of CD95 ligand (CD95L) or stimulated by anti-CD3 and -CD28 antibodies in the presence of recombinant CD95L had reduced activation and proliferation, whereas preactivated, CD95-sensitive T cells underwent apoptosis. Triggering of CD95 during T cell priming interfered with proximal T cell receptor signaling by inhibiting the recruitment of ζ-chain–associated protein of 70 kD, phospholipase-γ, and protein kinase C-θ into lipid rafts, thereby preventing their mutual tyrosine protein phosphorylation. Subsequently, Ca2+ mobilization and nuclear translocation of transcription factors NFAT, AP1, and NF-κB were strongly reduced, leading to impaired cytokine secretion. CD95-mediated inhibition of proliferation in naive T cells could not be reverted by the addition of exogenous interleukin-2 and T cells primed by CD95 co-stimulation remained partially unresponsive upon secondary T cell stimulation. HIV infection induced CD95L expression in primary human antigeen-presenting cells, and thereby suppressed T cell activation, suggesting that CD95/CD95L-mediated silencing of T cell activation represents a novel mechanism of immune evasion.
Collapse
|