1
|
Ahmad S, Single S, Liu Y, Hough KP, Wang Y, Thannickal VJ, Athar M, Goliwas KF, Deshane JS. Heavy Metal Exposure-Mediated Dysregulation of Sphingolipid Metabolism. Antioxidants (Basel) 2024; 13:978. [PMID: 39199224 DOI: 10.3390/antiox13080978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Exposure to heavy metals (HMs) is often associated with inflammation and cell death, exacerbating respiratory diseases including asthma. Most inhaled particulate HM exposures result in the deposition of HM-bound fine particulate matter, PM2.5, in pulmonary cell populations. While localized high concentrations of HMs may be a causative factor, existing studies have mostly evaluated the effects of systemic or low-dose chronic HM exposures. This report investigates the impact of local high concentrations of specific HMs (NaAsO2, MnCl2, and CdCl2) on sphingolipid homeostasis and oxidative stress, as both play a role in mediating responses to HM exposure and have been implicated in asthma. Utilizing an in vitro model system and three-dimensional ex vivo human tissue models, we evaluated the expression of enzymatic regulators of the salvage, recycling, and de novo synthesis pathways of sphingolipid metabolism, and observed differential modulation in these enzymes between HM exposures. Sphingolipidomic analyses of specific HM-exposed cells showed increased levels of anti-apoptotic sphingolipids and reduced pro-apoptotic sphingolipids, suggesting activation of the salvage and de novo synthesis pathways. Differential sphingolipid regulation was observed within HM-exposed lung tissues, with CdCl2 exposure and NaAsO2 exposure activating the salvage and de novo synthesis pathway, respectively. Additionally, using spatial transcriptomics and quantitative real-time PCR, we identified HM exposure-induced transcriptomic signatures of oxidative stress in epithelial cells and human lung tissues.
Collapse
Affiliation(s)
- Shaheer Ahmad
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Sierra Single
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Yuelong Liu
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Kenneth P Hough
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Yong Wang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Victor J Thannickal
- John W. Deming Department of Medicine, Tulane University School of Medicine and Southeast Veterans Healthcare System, New Orleans, LA 70119-6535, USA
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kayla F Goliwas
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Jessy S Deshane
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| |
Collapse
|
2
|
Jordan J, Levy JH, Gonzalez-Estrada A. Perioperative anaphylaxis: updates on pathophysiology. Curr Opin Allergy Clin Immunol 2024; 24:183-188. [PMID: 38743470 DOI: 10.1097/aci.0000000000000994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
PURPOSE OF REVIEW Perioperative anaphylaxis has historically been attributed to IgE/FcεRI-mediated reactions; there is now recognition of allergic and nonallergic triggers encompassing various reactions beyond IgE-mediated responses. This review aims to present recent advancements in knowledge regarding the mechanisms and pathophysiology of perioperative anaphylaxis. RECENT FINDINGS Emerging evidence highlights the role of the mast-cell related G-coupled protein receptor X2 pathway in direct mast cell degranulation, shedding light on previously unknown mechanisms. This pathway, alongside traditional IgE/FcεRI-mediated reactions, contributes to the complex nature of anaphylactic reactions. Investigations into the microbiota-anaphylaxis connection are ongoing, with potential implications for future treatment strategies. While serum tryptase levels serve as mast cell activation indicators, identifying triggers remains challenging. A range of mediators have been associated with anaphylaxis, including vasoactive peptides, proteases, lipid molecules, cytokines, chemokines, interleukins, complement components, and coagulation factors. SUMMARY Further understanding of clinical endotypes and the microenvironment where anaphylactic reactions unfold is essential for standardizing mediator testing and characterization in perioperative anaphylaxis. Ongoing research aims to elucidate the mechanisms, pathways, and mediators involved across multiple organ systems, including the cardiovascular, respiratory, and integumentary systems, which will be crucial for improving patient outcomes.
Collapse
Affiliation(s)
- Justin Jordan
- TMC Health Medical Education Program, Tucson, Arizona
| | - Jerrold H Levy
- Departments of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina
| | | |
Collapse
|
3
|
Carlucci CD, Hui Y, Chumanevich AP, Robida PA, Fuseler JW, Sajish M, Nagarkatti P, Nagarkatti M, Oskeritzian CA. Resveratrol Protects against Skin Inflammation through Inhibition of Mast Cell, Sphingosine Kinase-1, Stat3 and NF-κB p65 Signaling Activation in Mice. Int J Mol Sci 2023; 24:6707. [PMID: 37047680 PMCID: PMC10095068 DOI: 10.3390/ijms24076707] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
Inflammation is pathogenic to skin diseases, including atopic dermatitis (AD) and eczema. Treatment for AD remains mostly symptomatic with newer but costly options, tainted with adverse side effects. There is an unmet need for safe therapeutic and preventative strategies for AD. Resveratrol (R) is a natural compound known for its anti-inflammatory properties. However, animal and human R studies have yielded contrasting results. Mast cells (MCs) are innate immune skin-resident cells that initiate the development of inflammation and progression to overt disease. R's effects on MCs are also controversial. Using a human-like mouse model of AD development consisting of a single topical application of antigen ovalbumin (O) for 7 days, we previously established that the activation of MCs by a bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) initiated substantial skin remodeling compared to controls. Here, we show that daily R application normalized O-mediated epidermal thickening, ameliorated cell infiltration, and inhibited skin MC activation and chemokine expression. We unraveled R's multiple mechanisms of action, including decreased activation of the S1P-producing enzyme, sphingosine kinase 1 (SphK1), and of transcription factors Signal Transducer and Activator of Transcription 3 (Stat3) and NF-κBp65, involved in chemokine production. Thus, R may be poised for protection against MC-driven pathogenic skin inflammation.
Collapse
Affiliation(s)
- Christopher D Carlucci
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Yvonne Hui
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Alena P Chumanevich
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Piper A Robida
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - John W Fuseler
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Mathew Sajish
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Carole A Oskeritzian
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| |
Collapse
|
4
|
Baldo BA. Allergic and other adverse reactions to drugs used in anesthesia and surgery. ANESTHESIOLOGY AND PERIOPERATIVE SCIENCE 2023; 1:16. [PMCID: PMC10264870 DOI: 10.1007/s44254-023-00018-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/02/2023] [Accepted: 04/11/2023] [Indexed: 11/13/2023]
Abstract
The list of drugs patients may be exposed to during the perioperative and postoperative periods is potentially extensive. It includes induction agents, neuromuscular blocking drugs (NMBDs), opioids, antibiotics, sugammadex, colloids, local anesthetics, polypeptides, antifibrinolytic agents, heparin and related anticoagulants, blue dyes, chlorhexidine, and a range of other agents depending on several factors related to individual patients’ clinical condition and progress in the postoperative recovery period. To avoid poor or ultrarapid metabolizers to a particular drug (for example tramadol and codeine) or possible adverse drug reactions (ADRs), some drugs may need to be avoided during or after surgery. This will be the case for patients with a history of anaphylaxis or other adverse events/intolerances to a known drug. Other drugs may be ceased for a period before surgery, e.g., anticoagulants that increase the chance of bleeding; diuretics for patients with acute renal failure; antihypertensives relative to kidney injury after major vascular surgery; and serotonergic drugs that together with some opioids may rarely induce serotonin toxicity. Studies of germline variations shown by genotyping and phenotyping to identify a predisposition of genetic factors to ADRs offer an increasingly important approach to individualize drug therapy. Studies of associations of human leukocyte antigen (HLA) genes with some serious delayed immune-mediated reactions are ongoing and variations of drug-metabolizing cytochrome CYP450 enzymes, P-glycoprotein, and catechol-O -methyltransferase show promise for the assessment of ADRs and non-responses to drugs, particularly opioids and other analgesics. Surveys of ADRs from an increasing number of institutions often cover small numbers of patients, are retrospective in nature, fail to clearly identify culprit drugs, and do not adequately distinguish immune-mediated from non-immune-mediated anaphylactoid reactions. From the many surveys undertaken, the large list of agents identified during and after anesthesia and surgery are examined for their ADR involvement. Drugs are classified into those most often involved, (NMBD and antibiotics); drugs that are becoming more frequently implicated, namely antibiotics (particularly teicoplanin), and blue dyes; those becoming less frequently involved; and drugs more rarely involved in perioperative, and postoperative adverse reactions but still important and necessary to keep in mind for the occasional potential sensitive patient. Clinicians should be aware of the similarities between drug-induced true allergic type I IgE/FcεRI- and pseudoallergic MRGPRX2-mediated ADRs, the clinical features of each, and their distinguishing characteristics. Procedures for identifying MRGPRX2 agonists and diagnosing and distinguishing pseudoallergic from allergic reaction mechanisms are discussed.
Collapse
Affiliation(s)
- Brian A. Baldo
- Molecular Immunology Unit, Kolling Institute of Medical Research, Royal North Shore Hospital of Sydney, St Leonards, Australia
- Department of Medicine, University of Sydney, Sydney, NSW Australia
- Lindfield, Australia
| |
Collapse
|
5
|
Jo H, Shim K, Jeoung D. The Crosstalk between FcεRI and Sphingosine Signaling in Allergic Inflammation. Int J Mol Sci 2022; 23:ijms232213892. [PMID: 36430378 PMCID: PMC9695510 DOI: 10.3390/ijms232213892] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Sphingolipid molecules have recently attracted attention as signaling molecules in allergic inflammation diseases. Sphingosine-1-phosphate (S1P) is synthesized by two isoforms of sphingosine kinases (SPHK 1 and SPHK2) and is known to be involved in various cellular processes. S1P levels reportedly increase in allergic inflammatory diseases, such as asthma and anaphylaxis. FcεRI signaling is necessary for allergic inflammation as it can activate the SPHKs and increase the S1P level; once S1P is secreted, it can bind to the S1P receptors (S1PRs). The role of S1P signaling in various allergic diseases is discussed. Increased levels of S1P are positively associated with asthma and anaphylaxis. S1P can either induce or suppress allergic skin diseases in a context-dependent manner. The crosstalk between FcεRI and S1P/SPHK/S1PRs is discussed. The roles of the microRNAs that regulate the expression of the components of S1P signaling in allergic inflammatory diseases are also discussed. Various reports suggest the role of S1P in FcεRI-mediated mast cell (MC) activation. Thus, S1P/SPHK/S1PRs signaling can be the target for developing anti-allergy drugs.
Collapse
|
6
|
Masuda-Kuroki K, Di Nardo A. Sphingosine 1-Phosphate Signaling at the Skin Barrier Interface. BIOLOGY 2022; 11:biology11060809. [PMID: 35741330 PMCID: PMC9219813 DOI: 10.3390/biology11060809] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/14/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a product of membrane sphingolipid metabolism. S1P is secreted and acts via G-protein-coupled receptors, S1PR1-5, and is involved in diverse cellular functions, including cell proliferation, immune suppression, and cardiovascular functions. Recent studies have shown that the effects of S1P signaling are extended further by coupling the different S1P receptors and their respective downstream signaling pathways. Our group has recently reported that S1P inhibits cell proliferation and induces differentiation in human keratinocytes. There is a growing understanding of the connection between S1P signaling, skin barrier function, and skin diseases. For example, the activation of S1PR1 and S1PR2 during bacterial invasion regulates the synthesis of inflammatory cytokines in human keratinocytes. Moreover, S1P-S1PR2 signaling is involved in the production of inflammatory cytokines and can be triggered by epidermal mechanical stress and bacterial invasion. This review highlights how S1P affects human keratinocyte proliferation, differentiation, immunoreaction, and mast cell immune response, in addition to its effects on the skin barrier interface. Finally, studies targeting S1P-S1PR signaling involved in inflammatory skin diseases are also presented.
Collapse
|
7
|
Sasset L, Di Lorenzo A. Sphingolipid Metabolism and Signaling in Endothelial Cell Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:87-117. [PMID: 35503177 DOI: 10.1007/978-981-19-0394-6_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The endothelium, inner layer of blood vessels, constitutes a metabolically active paracrine, endocrine, and autocrine organ, able to sense the neighboring environment and exert a variety of biological functions important to preserve the health of vasculature, tissues, and organs. Sphingolipids are both fundamental structural components of the eukaryotic membranes and signaling molecules regulating a variety of biological functions. Ceramide and sphingosine-1-phosphate (S1P), bioactive sphingolipids, have emerged as important regulators of cardiovascular functions in health and disease. In this review we discuss recent insights into the role of ceramide and S1P biosynthesis and signaling in regulating endothelial cell functions, in health and diseases. We also highlight advances into the mechanisms regulating serine palmitoyltransferase, the first and rate-limiting enzyme of de novo sphingolipid biosynthesis, with an emphasis on its inhibitors, ORMDL and NOGO-B. Understanding the molecular mechanisms regulating the sphingolipid de novo biosynthesis may provide the foundation for therapeutic modulation of this pathway in a variety of conditions, including cardiovascular diseases, associated with derangement of this pathway.
Collapse
Affiliation(s)
- Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Rohrhofer J, Zwirzitz B, Selberherr E, Untersmayr E. The Impact of Dietary Sphingolipids on Intestinal Microbiota and Gastrointestinal Immune Homeostasis. Front Immunol 2021; 12:635704. [PMID: 34054805 PMCID: PMC8160510 DOI: 10.3389/fimmu.2021.635704] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
The large surfaces of gastrointestinal (GI) organs are well adapted to their diverse tasks of selective nutritional uptake and defense against the external environment. To maintain a functional balance, a vast number of immune cells is located within the mucosa. A strictly regulated immune response is required to impede constant inflammation and to maintain barrier function. An increasing prevalence of GI diseases has been reported in Western societies over the past decades. This surge in GI disorders has been linked to dietary changes followed by an imbalance of the gut microbiome, leading to a chronic, low grade inflammation of the gut epithelium. To counteract the increasing health care costs associated with diseases, it is paramount to understand the mechanisms driving immuno-nutrition, the associations between nutritional compounds, the commensal gut microbiota, and the host immune response. Dietary compounds such as lipids, play a central role in GI barrier function. Bioactive sphingolipids (SLs), e.g. sphingomyelin (SM), sphingosine (Sph), ceramide (Cer), sphingosine-1- phosphate (S1P) and ceramide-1-phosphate (C1P) may derive from dietary SLs ingested through the diet. They are not only integral components of cell membranes, they additionally modulate cell trafficking and are precursors for mediators and second messenger molecules. By regulating intracellular calcium levels, cell motility, cell proliferation and apoptosis, SL metabolites have been described to influence GI immune homeostasis positively and detrimentally. Furthermore, dietary SLs are suggested to induce a shift in the gut microbiota. Modes of action range from competing with the commensal bacteria for intestinal cell attachment to prevention from pathogen invasion by regulating innate and immediate defense mechanisms. SL metabolites can also be produced by gut microorganisms, directly impacting host metabolic pathways. This review aims to summarize recent findings on SL signaling and functional variations of dietary SLs. We highlight novel insights in SL homeostasis and SL impact on GI barrier function, which is directly linked to changes of the intestinal microbiota. Knowledge gaps in current literature will be discussed to address questions relevant for understanding the pivotal role of dietary SLs on chronic, low grade inflammation and to define a balanced and healthy diet for disease prevention and treatment.
Collapse
Affiliation(s)
- Johanna Rohrhofer
- Gastrointestinal Immunology Group, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Benjamin Zwirzitz
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Evelyne Selberherr
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Eva Untersmayr
- Gastrointestinal Immunology Group, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Bugajev V, Halova I, Demkova L, Cernohouzova S, Vavrova P, Mrkacek M, Utekal P, Draberova L, Kuchar L, Schuster B, Draber P. ORMDL2 Deficiency Potentiates the ORMDL3-Dependent Changes in Mast Cell Signaling. Front Immunol 2021; 11:591975. [PMID: 33643282 PMCID: PMC7905224 DOI: 10.3389/fimmu.2020.591975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
The systemic anaphylactic reaction is a life-threatening allergic response initiated by activated mast cells. Sphingolipids are an essential player in the development and attenuation of this response. De novo synthesis of sphingolipids in mammalian cells is inhibited by the family of three ORMDL proteins (ORMDL1, 2, and 3). However, the cell and tissue-specific functions of ORMDL proteins in mast cell signaling are poorly understood. This study aimed to determine cross-talk of ORMDL2 and ORMDL3 proteins in IgE-mediated responses. To this end, we prepared mice with whole-body knockout (KO) of Ormdl2 and/or Ormdl3 genes and studied their role in mast cell-dependent activation events in vitro and in vivo. We found that the absence of ORMDL3 in bone marrow-derived mast cells (BMMCs) increased the levels of cellular sphingolipids. Such an increase was further raised by simultaneous ORMDL2 deficiency, which alone had no effect on sphingolipid levels. Cells with double ORMDL2 and ORMDL3 KO exhibited increased intracellular levels of sphingosine-1-phosphate (S1P). Furthermore, we found that concurrent ORMDL2 and ORMDL3 deficiency increased IκB-α phosphorylation, degranulation, and production of IL-4, IL-6, and TNF-α cytokines in antigen-activated mast cells. Interestingly, the chemotaxis towards antigen was increased in all mutant cell types analyzed. Experiments in vivo showed that passive cutaneous anaphylaxis (PCA), which is initiated by mast cell activation, was increased only in ORMDL2,3 double KO mice, supporting our in vitro observations with mast cells. On the other hand, ORMDL3 KO and ORMDL2,3 double KO mice showed faster recovery from passive systemic anaphylaxis, which could be mediated by increased levels of blood S1P presented in such mice. Our findings demonstrate that Ormdl2 deficiency potentiates the ORMDL3-dependent changes in mast cell signaling.
Collapse
Affiliation(s)
- Viktor Bugajev
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Livia Demkova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Sara Cernohouzova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Petra Vavrova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Michal Mrkacek
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Pavol Utekal
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Lubica Draberova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Ladislav Kuchar
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Björn Schuster
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
10
|
Murine platelet production is suppressed by S1P release in the hematopoietic niche, not facilitated by blood S1P sensing. Blood Adv 2020; 3:1702-1713. [PMID: 31171507 DOI: 10.1182/bloodadvances.2019031948] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023] Open
Abstract
The bioactive lipid mediator sphingosine 1-phosphate (S1P) was recently assigned critical roles in platelet biology: whereas S1P1 receptor-mediated S1P gradient sensing was reported to be essential for directing proplatelet extensions from megakaryocytes (MKs) toward bone marrow sinusoids, MK sphingosine kinase 2 (Sphk2)-derived S1P was reported to further promote platelet shedding through receptor-independent intracellular actions, and platelet aggregation through S1P1 Yet clinical use of S1P pathway modulators including fingolimod has not been associated with risk of bleeding or thrombosis. We therefore revisited the role of S1P in platelet biology in mice. Surprisingly, no reduction in platelet counts was observed when the vascular S1P gradient was ablated by impairing S1P provision to plasma or S1P degradation in interstitial fluids, nor when gradient sensing was impaired by S1pr1 deletion selectively in MKs. Moreover, S1P1 expression and signaling were both undetectable in mature MKs in situ, and MK S1pr1 deletion did not affect platelet aggregation or spreading. When S1pr1 deletion was induced in hematopoietic progenitor cells, platelet counts were instead significantly elevated. Isolated global Sphk2 deficiency was associated with thrombocytopenia, but this was not replicated by MK-restricted Sphk2 deletion and was reversed by compound deletion of either Sphk1 or S1pr2, suggesting that this phenotype arises from increased S1P export and S1P2 activation secondary to redistribution of sphingosine to Sphk1. Consistent with clinical observations, we thus observe no essential role for S1P1 in facilitating platelet production or activation. Instead, S1P restricts megakaryopoiesis through S1P1, and can further suppress thrombopoiesis through S1P2 when aberrantly secreted in the hematopoietic niche.
Collapse
|
11
|
Ishay Y, Nachman D, Khoury T, Ilan Y. The role of the sphingolipid pathway in liver fibrosis: an emerging new potential target for novel therapies. Am J Physiol Cell Physiol 2020; 318:C1055-C1064. [PMID: 32130072 DOI: 10.1152/ajpcell.00003.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sphingolipids (SL) are a family of bioactive lipids and a major cellular membrane structural component. SLs include three main compounds: ceramide (Cer), sphingosine (Sp), and sphingosine-1-phosphate (S-1P), all of which have emerging roles in biological functions in cells, especially in the liver. They are under investigation in various liver diseases, including cirrhosis and end-stage liver disease. In this review, we provide an overview on the role of SLs in liver pathobiology and focus on their potential role in the development of hepatic fibrosis. We describe recent evidence and suggest SLs are a promising potential therapeutic target for the treatment of liver disease and fibrosis.
Collapse
Affiliation(s)
- Yuval Ishay
- Department of Internal Medicine A, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dean Nachman
- Department of Internal Medicine A, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tawfik Khoury
- Gastroenterology and Liver Units, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
12
|
Kharel Y, Huang T, Salamon A, Harris TE, Santos WL, Lynch KR. Mechanism of sphingosine 1-phosphate clearance from blood. Biochem J 2020; 477:925-935. [PMID: 32065229 PMCID: PMC7059866 DOI: 10.1042/bcj20190730] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/27/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023]
Abstract
The interplay of sphingosine 1-phosphate (S1P) synthetic and degradative enzymes as well as S1P exporters creates concentration gradients that are a fundamental to S1P biology. Extracellular S1P levels, such as in blood and lymph, are high relative to cellular S1P. The blood-tissue S1P gradient maintains endothelial integrity while local S1P gradients influence immune cell positioning. Indeed, the importance of S1P gradients was recognized initially when the mechanism of action of an S1P receptor agonist used as a medicine for multiple sclerosis was revealed to be inhibition of T-lymphocytes' recognition of the high S1P in efferent lymph. Furthermore, the increase in erythrocyte S1P in response to hypoxia influences oxygen delivery during high altitude acclimatization. However, understanding of how S1P gradients are maintained is incomplete. For example, S1P is synthesized but is only slowly metabolized by blood yet circulating S1P turns over quickly by an unknown mechanism. Prompted by the counterintuitive observation that blood S1P increases markedly in response to inhibition S1P synthesis (by sphingosine kinase 2 (SphK2)), we studied mice wherein several tissues were made deficient in either SphK2 or S1P degrading enzymes. Our data reveal a mechanism whereby S1P is de-phosphorylated at the hepatocyte surface and the resulting sphingosine is sequestered by SphK phosphorylation and in turn degraded by intracellular S1P lyase. Thus, we identify the liver as the primary site of blood S1P clearance and provide an explanation for the role of SphK2 in this process. Our discovery suggests a general mechanism whereby S1P gradients are shaped.
Collapse
Affiliation(s)
- Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, U.S.A
| | - Tao Huang
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, U.S.A
| | - Anita Salamon
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, U.S.A
| | - Thurl E. Harris
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, U.S.A
| | - Webster L. Santos
- Department of Chemistry and VT Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, U.S.A
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, U.S.A
| |
Collapse
|
13
|
Ishimaru K, Yoshioka K, Kano K, Kurano M, Saigusa D, Aoki J, Yatomi Y, Takuwa N, Okamoto Y, Proia RL, Takuwa Y. Sphingosine kinase-2 prevents macrophage cholesterol accumulation and atherosclerosis by stimulating autophagic lipid degradation. Sci Rep 2019; 9:18329. [PMID: 31797978 PMCID: PMC6892873 DOI: 10.1038/s41598-019-54877-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is the major cause of ischemic coronary heart diseases and characterized by the infiltration of cholesterol-accumulating macrophages in the vascular wall. Although sphingolipids are implicated in atherosclerosis as both membrane components and lipid mediators, the precise role of sphingolipids in atherosclerosis remains elusive. Here, we found that genetic deficiency of sphingosine kinase-2 (SphK2) but not SphK1 aggravates the formation of atherosclerotic lesions in mice with ApoE deficiency. Bone marrow chimaera experiments show the involvement of SphK2 expressed in bone marrow-derived cells. In macrophages, deficiency of SphK2, a major SphK isoform in this cell type, results in increases in cellular sphingosine and ceramides. SphK2-deficient macrophages have increases in lipid droplet-containing autophagosomes and autolysosomes and defective lysosomal degradation of lipid droplets via autophagy with an impaired luminal acidic environment and proteolytic activity in the lysosomes. Transgenic overexpression of SphK1 in SphK2-deficient mice rescued aggravation of atherosclerosis and abnormalities of autophagosomes and lysosomes in macrophages with reductions of sphingosine, suggesting at least partial overlapping actions of two SphKs. Taken together, these results indicate that SphK2 is required for autophagosome- and lysosome-mediated catabolism of intracellular lipid droplets to impede the development of atherosclerosis; therefore, SphK2 may be a novel target for treating atherosclerosis.
Collapse
Affiliation(s)
- Kazuhiro Ishimaru
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan.
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan
| | - Kuniyuki Kano
- Department of Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, 980-8578, Japan
| | - Makoto Kurano
- Department of Laboratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, 113-0033, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, 980-8573, Japan
| | - Junken Aoki
- Department of Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, 980-8578, Japan
| | - Yutaka Yatomi
- Department of Laboratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, 113-0033, Japan
| | - Noriko Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan.,Department of Health and Medical Sciences, Ishikawa Prefectural Nursing University, Kahoku, 929-1210, Japan
| | - Yasuo Okamoto
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, 20892, USA
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan.
| |
Collapse
|
14
|
Wang P, Yuan Y, Lin W, Zhong H, Xu K, Qi X. Roles of sphingosine-1-phosphate signaling in cancer. Cancer Cell Int 2019; 19:295. [PMID: 31807117 PMCID: PMC6857321 DOI: 10.1186/s12935-019-1014-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
The potent pleiotropic lipid mediator sphingosine-1-phosphate (S1P) participates in numerous cellular processes, including angiogenesis and cell survival, proliferation, and migration. It is formed by one of two sphingosine kinases (SphKs), SphK1 and SphK2. These enzymes largely exert their various biological and pathophysiological actions through one of five G protein-coupled receptors (S1PR1–5), with receptor activation setting in motion various signaling cascades. Considerable evidence has been accumulated on S1P signaling and its pathogenic roles in diseases, as well as on novel modulators of S1P signaling, such as SphK inhibitors and S1P agonists and antagonists. S1P and ceramide, composed of sphingosine and a fatty acid, are reciprocal cell fate regulators, and S1P signaling plays essential roles in several diseases, including inflammation, cancer, and autoimmune disorders. Thus, targeting of S1P signaling may be one way to block the pathogenesis and may be a therapeutic target in these conditions. Increasingly strong evidence indicates a role for the S1P signaling pathway in the progression of cancer and its effects. In the present review, we discuss recent progress in our understanding of S1P and its related proteins in cancer progression. Also described is the therapeutic potential of S1P receptors and their downstream signaling cascades as targets for cancer treatment.
Collapse
Affiliation(s)
- Peng Wang
- 1Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Yonghui Yuan
- 1Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China.,2Research and Academic Department, Cancer Hospital of China Medical University Liaoning Cancer Hospital & Institute, Shenyang, 110042 Liaoning China
| | - Wenda Lin
- 1Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Hongshan Zhong
- 1Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Ke Xu
- 1Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Xun Qi
- 1Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China
| |
Collapse
|
15
|
Jena PK, Sheng L, Mcneil K, Chau TQ, Yu S, Kiuru M, Fung MA, Hwang ST, Wan YJY. Long-term Western diet intake leads to dysregulated bile acid signaling and dermatitis with Th2 and Th17 pathway features in mice. J Dermatol Sci 2019; 95:13-20. [PMID: 31213388 DOI: 10.1016/j.jdermsci.2019.05.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 05/09/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Dietary interventions are implicated in the development of atopic dermatitis, psoriasis, and acne. OBJECTIVE To investigate the effect of diet and the bile acid (BA) receptors, such as TGR5 (Takeda G protein receptor 5) and S1PR2 (sphingosine-1-phosphate receptor 2) in the development of dermatitis. METHODS C57BL/6 mice were fed a control diet (CD) or Western diet (WD) since weaning until they were 10 months old followed by analyzing histology, gene expression, and BA profiling. RESULTS Mice developed dermatitis as they aged and the incidence was higher in females than males. Additionally, WD intake substantially increased the incidence of dermatitis. Cutaneous antimicrobial peptide genesS100A8, S100A9, and Defb4 were reduced in WD-fed mice, but increased when mice developed skin lesions. In addition, Tgr5 and TGR5-regulated Dio2 and Nos3 were reduced in WD intake but induced in dermatitic lesions. Trpa1 and Trpv1, which mediate itch, were also increased in dermatitic lesions. The expression of S1pr2 and genes encoding sphingosine kinases, S1P phosphatases, binding protein, and transporter were all reduced by WD intake but elevated in dermatitic lesions. Furthermore, dermatitis development increased total cutaneous BA with an altered profile, which may change TGR5 and S1PR2 activity. Moreover, supplementation with BA sequestrant cholestyramine reduced epidermal thickening as well as cutaneous inflammatory cytokines. CONCLUSION In summary, activation of TGR5 and S1PR2, which regulate itch, keratinocyte proliferation, metabolism, and inflammation, may contribute to WD-exacerbated dermatitis with Th2 and Th17 features. In addition, elevated total BA play a significant role in inducing dermatitis and cutaneous inflammation.
Collapse
Affiliation(s)
- Prasant Kumar Jena
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Lili Sheng
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Kyle Mcneil
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Thinh Q Chau
- Department of Dermatology, University of California, Davis, Sacramento, CA, 95817, USA
| | - Sebastian Yu
- Department of Dermatology, University of California, Davis, Sacramento, CA, 95817, USA
| | - Maija Kiuru
- Department of Dermatology, University of California, Davis, Sacramento, CA, 95817, USA
| | - Maxwell A Fung
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, 95817, USA; Department of Dermatology, University of California, Davis, Sacramento, CA, 95817, USA
| | - Samuel T Hwang
- Department of Dermatology, University of California, Davis, Sacramento, CA, 95817, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
16
|
Role of Bioactive Sphingolipids in Inflammation and Eye Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1161:149-167. [PMID: 31562629 DOI: 10.1007/978-3-030-21735-8_14] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation is a common underlying factor in a diversity of ocular diseases, ranging from macular degeneration, autoimmune uveitis, glaucoma, diabetic retinopathy and microbial infection. In addition to the variety of known cellular mediators of inflammation, such as cytokines, chemokines and lipid mediators, there is now considerable evidence that sphingolipid metabolites also play a central role in the regulation of inflammatory pathways. Various sphingolipid metabolites, such as ceramide (Cer), ceramide-1-phosphate (C1P), sphingosine-1-phosphate (S1P), and lactosylceramide (LacCer) can contribute to ocular inflammatory diseases through multiple pathways. For example, inflammation generates Cer from sphingomyelins (SM) in the plasma membrane, which induces death receptor ligand formation and leads to apoptosis of retinal pigment epithelial (RPE) and photoreceptor cells. Inflammatory stress by reactive oxygen species leads to LacCer accumulation and S1P secretion and induces proliferation of retinal endothelial cells and eventual formation of new vessels. In sphingolipid/lysosomal storage disorders, sphingolipid metabolites accumulate in lysosomes and can cause ocular disorders that have an inflammatory etiology. Sphingolipid metabolites activate complement factors in the immune-response mediated pathogenesis of macular degeneration. These examples highlight the integral association between sphingolipids and inflammation in ocular diseases.
Collapse
|
17
|
Hasanifard L, Sheervalilou R, Majidinia M, Yousefi B. New insights into the roles and regulation of SphK2 as a therapeutic target in cancer chemoresistance. J Cell Physiol 2018; 234:8162-8181. [PMID: 30456838 DOI: 10.1002/jcp.27612] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/24/2018] [Indexed: 12/21/2022]
Abstract
Chemoresistance is a complicated process developed by most cancers and accounts for the majority of relapse and metastasis in cancer. The main mechanisms of chemoresistance phenotype include increased expression and/or activated drug efflux pumps, altered DNA repair, altered metabolism of therapeutics as well as impaired apoptotic signaling pathways. Aberrant sphingolipid signaling has also recently received considerable attention in chemoresistance. Sphingolipid metabolites regulate main biological processes such as apoptosis, cell survival, proliferation, and differentiation. Two sphingosine kinases, SphK1 and SphK2, convert sphingosine to sphingosine-1-phosphate, an antiapoptotic bioactive lipid mediator. Numerous evidence has revealed the involvement of activated SphK1 in tumorigenesis and resistance, however, contradictory results have been found for the role of SphK2 in these functions. In some studies, overexpression of SphK2 suppressed cell growth and induced apoptosis. In contrast, some others have shown cell proliferation and tumor promotion effect for SphK2. Our understanding of the role of SphK2 in cancer does not have a sufficient integrity. The main focus of this review will be on the re-evaluation of the role of SphK2 in cell death and chemoresistance in light of our new understanding of molecular targeted therapy. We will also highlight the connections between SphK2 and the DNA damage response. Finally, we will provide our insight into the regulatory mechanisms of SphKs by two main categories, micro and long, noncoding RNAs as the novel players of cancer chemoresistance.
Collapse
Affiliation(s)
- Leili Hasanifard
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghayeh Sheervalilou
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
S1P₄ Regulates Passive Systemic Anaphylaxis in Mice but Is Dispensable for Canonical IgE-Mediated Responses in Mast Cells. Int J Mol Sci 2018; 19:ijms19051279. [PMID: 29693558 PMCID: PMC5983835 DOI: 10.3390/ijms19051279] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
Mast cells are key players in the development of inflammatory allergic reactions. Cross-linking of the high-affinity receptor for IgE (FcεRI) on mast cells leads to the generation and secretion of the sphingolipid mediator, sphingosine-1-phosphate (S1P) which is able, in turn, to transactivate its receptors on mast cells. Previous reports have identified the expression of two of the five receptors for S1P on mast cells, S1P1 and S1P2, with functions in FcεRI-mediated chemotaxis and degranulation, respectively. Here, we show that cultured mouse mast cells also express abundant message for S1P4. Genetic deletion of S1pr4 did not affect the differentiation of bone marrow progenitors into mast cells or the proliferation of mast cells in culture. A comprehensive characterization of IgE-mediated responses in S1P4-deficient bone marrow-derived and peritoneal mouse mast cells indicated that this receptor is dispensable for mast cell degranulation, cytokine/chemokine production and FcεRI-mediated chemotaxis in vitro. However, interleukin-33 (IL-33)-mediated enhancement of IgE-induced degranulation was reduced in S1P4-deficient peritoneal mast cells, revealing a potential negative regulatory role for S1P4 in an IL-33-rich environment. Surprisingly, genetic deletion of S1pr4 resulted in exacerbation of passive systemic anaphylaxis to IgE/anti-IgE in mice, a phenotype likely related to mast cell-extrinsic influences, such as the high circulating levels of IgE in these mice which increases FcεRI expression and consequently the extent of the response to FcεRI engagement. Thus, we provide evidence that S1P4 modulates anaphylaxis in an unexpected manner that does not involve regulation of mast cell responsiveness to IgE stimulation.
Collapse
|
19
|
Bandara G, Muñoz-Cano R, Tobío A, Yin Y, Komarow HD, Desai A, Metcalfe DD, Olivera A. Targeting Sphingosine Kinase Isoforms Effectively Reduces Growth and Survival of Neoplastic Mast Cells With D816V-KIT. Front Immunol 2018; 9:631. [PMID: 29643855 PMCID: PMC5883065 DOI: 10.3389/fimmu.2018.00631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/13/2018] [Indexed: 01/02/2023] Open
Abstract
Mastocytosis is a disorder resulting from an abnormal mast cell (MC) accumulation in tissues that is often associated with the D816V mutation in KIT, the tyrosine kinase receptor for stem cell factor. Therapies available to treat aggressive presentations of mastocytosis are limited, thus exploration of novel pharmacological targets that reduce MC burden is desirable. Since increased generation of the lipid mediator sphingosine-1-phosphate (S1P) by sphingosine kinase (SPHK) has been linked to oncogenesis, we studied the involvement of the two SPHK isoforms (SPHK1 and SPHK2) in the regulation of neoplastic human MC growth. While SPHK2 inhibition prevented entry into the cell cycle in normal and neoplastic human MCs with minimal effect on cell survival, SPHK1 inhibition caused cell cycle arrest in G2/M and apoptosis, particularly in D816V-KIT MCs. This was mediated via activation of the DNA damage response (DDR) cascade, including phosphorylation of the checkpoint kinase 2 (CHK2), CHK2-mediated M-phase inducer phosphatase 3 depletion, and p53 activation. Combination treatment of SPHK inhibitors with KIT inhibitors showed greater growth inhibition of D816V-KIT MCs than either inhibitor alone. Furthermore, inhibition of SPHK isoforms reduced the number of malignant bone marrow MCs from patients with mastocytosis and the growth of D816V-KIT MCs in a xenograft mouse model. Our results reveal a role for SPHK isoforms in the regulation of growth and survival in normal and neoplastic MCs and suggest a regulatory function for SPHK1 in the DDR in MCs with KIT mutations. The findings also suggest that targeting the SPHK/S1P axis may provide an alternative to tyrosine kinase inhibitors, alone or in combination, for the treatment of aggressive mastocytosis and other hematological malignancies associated with the D816V-KIT mutation.
Collapse
Affiliation(s)
- Geethani Bandara
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rosa Muñoz-Cano
- Allergy Section, Pneumology Department, Hospital Clinic, ARADyAL, Instituto de Salud Carlos III, Barcelona, Spain
| | - Araceli Tobío
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yuzhi Yin
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Hirsh D Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Avanti Desai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
20
|
Redegeld FA, Yu Y, Kumari S, Charles N, Blank U. Non-IgE mediated mast cell activation. Immunol Rev 2018; 282:87-113. [DOI: 10.1111/imr.12629] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Frank A. Redegeld
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Yingxin Yu
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Sangeeta Kumari
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Nicolas Charles
- INSERM U1149; Centre de Recherche sur l'Inflammation; Paris France
- CNRS ERL8252; Paris France
- Université Paris-Diderot; Sorbonne Paris Cité; Faculté de Médecine; Site Xavier Bichat; Paris France
| | - Ulrich Blank
- INSERM U1149; Centre de Recherche sur l'Inflammation; Paris France
- CNRS ERL8252; Paris France
- Université Paris-Diderot; Sorbonne Paris Cité; Faculté de Médecine; Site Xavier Bichat; Paris France
- Inflamex Laboratory of Excellence; Paris France
| |
Collapse
|
21
|
Adamiak M, Chelvarajan L, Lynch KR, Santos WL, Abdel-Latif A, Ratajczak MZ. Mobilization studies in mice deficient in sphingosine kinase 2 support a crucial role of the plasma level of sphingosine-1-phosphate in the egress of hematopoietic stem progenitor cells. Oncotarget 2017; 8:65588-65600. [PMID: 29029455 PMCID: PMC5630355 DOI: 10.18632/oncotarget.19514] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 07/14/2017] [Indexed: 11/25/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid involved in cell signaling and, if released from cells, also plays a crucial role in regulating the trafficking of lympho-hematopoietic cells, including primitive hematopoietic stem/progenitor cells (HSPCs). It has been demonstrated that S1P chemoattracts HSPCs, and its level in peripheral blood creates a gradient directing egress of these cells during mobilization. In this paper we analyzed hematopoiesis in mice deficient in sphingosine kinase 2 (Sphk2-KO mice) and studied the effect of this mutation on plasma S1P levels. We found that Sphk2-KO mice have normal hematopoiesis, and, in contrast to Sphk1-KO mice, the circulating S1P level is highly elevated in these animals and correlates with the fact that HSPCs in Sphk2-KO animals, also in contrast to Sphk1-KO animals, show enhanced mobilization. These results were recapitulated in wild type (WT) animals employing an Sphk2 inhibitor. We also administered an inhibitor of the S1P-degrading enzyme S1P lyase, known as tetrahydroxybutylimidazole (THI), to WT mice and observed that this resulted in an increase in S1P level in PB and enhanced mobilization of HSPCs. In sum, our results support a crucial role for S1P gradients in blood plasma in the mobilization process and indicate that small-molecule inhibitors of Sphk2 and Sgpl1 could be employed as mobilization-facilitating compounds. At the same time, further studies are needed to explain the unexpected effect of Sphk2 inhibition on increasing S1P levels in plasma.
Collapse
Affiliation(s)
- Mateusz Adamiak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | - Lakshman Chelvarajan
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Kevin R. Lynch
- Department of Pharmacology University of Virginia, Charlottesville, VA, USA
| | - Webster L. Santos
- Department of Chemistry, Center for Drug Discovery, Virginia Tech, Blacksburg, VA, USA
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Mariusz Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
22
|
Reber LL, Hernandez JD, Galli SJ. The pathophysiology of anaphylaxis. J Allergy Clin Immunol 2017; 140:335-348. [PMID: 28780941 PMCID: PMC5657389 DOI: 10.1016/j.jaci.2017.06.003] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 01/14/2023]
Abstract
Anaphylaxis is a severe systemic hypersensitivity reaction that is rapid in onset; characterized by life-threatening airway, breathing, and/or circulatory problems; and usually associated with skin and mucosal changes. Because it can be triggered in some persons by minute amounts of antigen (eg, certain foods or single insect stings), anaphylaxis can be considered the most aberrant example of an imbalance between the cost and benefit of an immune response. This review will describe current understanding of the immunopathogenesis and pathophysiology of anaphylaxis, focusing on the roles of IgE and IgG antibodies, immune effector cells, and mediators thought to contribute to examples of the disorder. Evidence from studies of anaphylaxis in human subjects will be discussed, as well as insights gained from analyses of animal models, including mice genetically deficient in the antibodies, antibody receptors, effector cells, or mediators implicated in anaphylaxis and mice that have been "humanized" for some of these elements. We also review possible host factors that might influence the occurrence or severity of anaphylaxis. Finally, we will speculate about anaphylaxis from an evolutionary perspective and argue that, in the context of severe envenomation by arthropods or reptiles, anaphylaxis might even provide a survival advantage.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Immunology, Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Paris, France; Institut National de la Santé et de la Recherche Médicale, Paris, France; Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Joseph D Hernandez
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, Stanford University School of Medicine, Stanford, Calif
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
23
|
Saluja R, Kumar A, Jain M, Goel SK, Jain A. Role of Sphingosine-1-Phosphate in Mast Cell Functions and Asthma and Its Regulation by Non-Coding RNA. Front Immunol 2017; 8:587. [PMID: 28588581 PMCID: PMC5439123 DOI: 10.3389/fimmu.2017.00587] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/03/2017] [Indexed: 01/07/2023] Open
Abstract
Sphingolipid metabolites are emerging as important signaling molecules in allergic diseases specifically asthma. One of the sphingolipid metabolite, sphingosine-1-phosphate (S1P), is involved in cell differentiation, proliferation, survival, migration, and angiogenesis. In the allergic diseases, alteration of S1P levels influences the differentiation and responsiveness of mast cells (MCs). S1P is synthesized by two sphingosine kinases (SphKs), sphingosine kinase 1, and sphingosine kinase 2. Engagement of IgE to the FcεRI receptor induces the activation of both the SphKs and generates S1P. Furthermore, SphKs are also essential to FcεRI-mediated MC activation. Activated MCs export S1P into the extracellular space and causes inflammatory response and tissue remodeling. S1P signaling has dual role in allergic responses. Activation of SphKs and secretion of S1P are required for MC activation; however, S1P signaling plays a vital role in the recovery from anaphylaxis. Several non-coding RNAs have been shown to play a crucial role in controlling the MC-associated inflammatory and allergic responses. Thus, S1P signaling pathway and its regulation by non-coding RNA could be explored as an exciting potential therapeutic target for asthma and other MC-associated diseases.
Collapse
Affiliation(s)
- Rohit Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| | - Manju Jain
- Centre for Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda, India
| | - Sudhir K Goel
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| | - Aklank Jain
- Centre for Animal Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
24
|
Abstract
Vertebrates are endowed with a closed circulatory system, the evolution of which required novel structural and regulatory changes. Furthermore, immune cell trafficking paradigms adapted to the barriers imposed by the closed circulatory system. How did such changes occur mechanistically? We propose that spatial compartmentalization of the lipid mediator sphingosine 1-phosphate (S1P) may be one such mechanism. In vertebrates, S1P is spatially compartmentalized in the blood and lymphatic circulation, thus comprising a sharp S1P gradient across the endothelial barrier. Circulatory S1P has critical roles in maturation and homeostasis of the vascular system as well as in immune cell trafficking. Physiological functions of S1P are tightly linked to shear stress, the key biophysical stimulus from blood flow. Thus, circulatory S1P confinement could be a primordial strategy of vertebrates in the development of a closed circulatory system. This review discusses the cellular and molecular basis of the S1P gradients and aims to interpret its physiological significance as a key feature of the closed circulatory system.
Collapse
Affiliation(s)
- Keisuke Yanagida
- Vascular Biology Program, Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts 02115; ,
| | - Timothy Hla
- Vascular Biology Program, Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts 02115; ,
| |
Collapse
|
25
|
Roviezzo F, Sorrentino R, Iacono VM, Brancaleone V, Terlizzi M, Riemma MA, Bertolino A, Rossi A, Matteis M, Spaziano G, Pinto A, D'Agostino B, Cirino G. Disodium cromoglycate inhibits asthma-like features induced by sphingosine-1-phosphate. Pharmacol Res 2016; 113:626-635. [PMID: 27713021 DOI: 10.1016/j.phrs.2016.09.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/28/2016] [Accepted: 09/14/2016] [Indexed: 11/17/2022]
Abstract
Compelling evidence suggests the involvement of sphingosine-1-phosphate (S1P) in the pathogenesis of asthma. The systemic administration of S1P causes asthma like features in the mouse involving mast cells. In this study we investigated whether disodium cromoglycate (DSCG), administered as a preventative treatment as in human therapy, could affect S1P effects on airways. BALB/c mice, treated with DSCG, received subcutaneous administration of S1P. Bronchi and pulmonary tissues were collected and functional, molecular and cellular studies were performed. DSCG inhibited S1P-induced airway hyper-reactivity as well as pulmonary inflammation. DSCG decreased the recruitment of solely mast cells and B cells in the lung. IgE serum levels, prostaglandin D2, mucus production and IL-13 were also reduced when mice were pretreated with DSCG. S1P induced pulmonary expression of CD23 on T and B cells, that was reversed by DSCG. Conversely, S1P failed to upregulate CD23 in mast cell-deficient Kit W-sh/W-sh mice. In conclusion we have shown that DSCG inhibits S1P-induced asthma like features in the mouse. This beneficial effect is due to a regulatory action on mast cell activity, and in turn to an inhibition of IgE-dependent T and B cells responses.
Collapse
Affiliation(s)
| | | | | | | | - Michela Terlizzi
- Department of Pharmacy (DIFARMA), University of Salerno, Salerno, Italy
| | | | - Antonio Bertolino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Antonietta Rossi
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Maria Matteis
- Department of Experimental Medicine L. Donatelli, Second University of Naples, Naples, Italy
| | - Giuseppe Spaziano
- Department of Experimental Medicine L. Donatelli, Second University of Naples, Naples, Italy
| | - Aldo Pinto
- Department of Pharmacy (DIFARMA), University of Salerno, Salerno, Italy
| | - Bruno D'Agostino
- Department of Experimental Medicine L. Donatelli, Second University of Naples, Naples, Italy.
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
26
|
Pitman MR, Costabile M, Pitson SM. Recent advances in the development of sphingosine kinase inhibitors. Cell Signal 2016; 28:1349-1363. [DOI: 10.1016/j.cellsig.2016.06.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
|
27
|
Gazit SL, Mariko B, Thérond P, Decouture B, Xiong Y, Couty L, Bonnin P, Baudrie V, Le Gall SM, Dizier B, Zoghdani N, Ransinan J, Hamilton JR, Gaussem P, Tharaux PL, Chun J, Coughlin SR, Bachelot-Loza C, Hla T, Ho-Tin-Noé B, Camerer E. Platelet and Erythrocyte Sources of S1P Are Redundant for Vascular Development and Homeostasis, but Both Rendered Essential After Plasma S1P Depletion in Anaphylactic Shock. Circ Res 2016; 119:e110-26. [PMID: 27582371 DOI: 10.1161/circresaha.116.308929] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/30/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Sphingosine-1-phosphate (S1P) signaling is essential for vascular development and postnatal vascular homeostasis. The relative importance of S1P sources sustaining these processes remains unclear. OBJECTIVE To address the level of redundancy in bioactive S1P provision to the developing and mature vasculature. METHODS AND RESULTS S1P production was selectively impaired in mouse platelets, erythrocytes, endothelium, or smooth muscle cells by targeted deletion of genes encoding sphingosine kinases -1 and -2. S1P deficiency impaired aggregation and spreading of washed platelets and profoundly reduced their capacity to promote endothelial barrier function ex vivo. However, and in contrast to recent reports, neither platelets nor any other source of S1P was essential for vascular development, vascular integrity, or hemostasis/thrombosis. Yet rapid and profound depletion of plasma S1P during systemic anaphylaxis rendered both platelet- and erythrocyte-derived S1P essential for survival, with a contribution from blood endothelium observed only in the absence of circulating sources. Recovery was sensitive to aspirin in mice with but not without platelet S1P, suggesting that platelet activation and stimulus-response coupling is needed. S1P deficiency aggravated vasoplegia in this model, arguing a vital role for S1P in maintaining vascular resistance during recovery from circulatory shock. Accordingly, the S1P2 receptor mediated most of the survival benefit of S1P, whereas the endothelial S1P1 receptor was dispensable for survival despite its importance for maintaining vascular integrity. CONCLUSIONS Although source redundancy normally secures essential S1P signaling in developing and mature blood vessels, profound depletion of plasma S1P renders both erythrocyte and platelet S1P pools necessary for recovery and high basal plasma S1P levels protective during anaphylactic shock.
Collapse
Affiliation(s)
- Salomé L Gazit
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Boubacar Mariko
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Patrice Thérond
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Benoit Decouture
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Yuquan Xiong
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Ludovic Couty
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Philippe Bonnin
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Véronique Baudrie
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Sylvain M Le Gall
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Blandine Dizier
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Nesrine Zoghdani
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Jessica Ransinan
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Justin R Hamilton
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Pascale Gaussem
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Pierre-Louis Tharaux
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Jerold Chun
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Shaun R Coughlin
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Christilla Bachelot-Loza
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Timothy Hla
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Benoit Ho-Tin-Noé
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.)
| | - Eric Camerer
- From the INSERM U970, Paris Cardiovascular Research Centre, 75015 Paris, France (S.L.G., B.M., L.C., V.B., S.M.L.G., N.Z., J.R., P.-L.T., E.C.); Université Sorbonne Paris Cité, Paris, France (S.L.G., B.M., B. Decouture, L.C., P.B., V.B., S.M.L.G., B. Dizier, N.Z., J.R., P.G., P.-L.T., C.B.-L., B.H.-T.-N., E.C.); AP-HP, Hôpital Bicêtre, Service de Biochimie, 94275 Le Kremlin Bicêtre, France (P.T.); Lip(Sys)2-Biochimie appliquée, Université Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France (P.T.); INSERM U1140, Faculté de Pharmacie, 75006 Paris, France (B. Decouture, B. Dizier, P.G., C.B.-L.); Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York (Y.X., T.H.); AP-HP, Hôpital Lariboisière, Physiologie Clinique-Explorations-Fonctionnelles, INSERM U965, 75010, Paris, France (P.B.); Australian Centre for Blood Diseases & Department of Clinical Haematology, Monash University, Melbourne, Australia (J.R.H.); AP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France (P.G.); Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.); Cardiovascular Research Institute, University of California, San Francisco (S.R.C.); and INSERM U698, 75018 Paris, France (B.H.-T.-N.).
| |
Collapse
|
28
|
Cantalupo A, Di Lorenzo A. S1P Signaling and De Novo Biosynthesis in Blood Pressure Homeostasis. J Pharmacol Exp Ther 2016; 358:359-70. [PMID: 27317800 PMCID: PMC4959106 DOI: 10.1124/jpet.116.233205] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/13/2016] [Indexed: 01/12/2023] Open
Abstract
Initially discovered as abundant components of eukaryotic cell membranes, sphingolipids are now recognized as important bioactive signaling molecules that modulate a variety of cellular functions, including those relevant to cancer and immunologic, inflammatory, and cardiovascular disorders. In this review, we discuss recent advances in our understanding of the role of sphingosine-1-phosphate (S1P) receptors in the regulation of vascular function, and focus on how de novo biosynthesized sphingolipids play a role in blood pressure homeostasis. The therapeutic potential of new drugs that target S1P signaling is also discussed.
Collapse
Affiliation(s)
- Anna Cantalupo
- Department of Pathology and Laboratory Medicine, Center for Vascular Biology, Weill Cornell Medicine, Cornell University, New York, New York
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Center for Vascular Biology, Weill Cornell Medicine, Cornell University, New York, New York
| |
Collapse
|
29
|
Nagahashi M, Yamada A, Miyazaki H, Allegood JC, Tsuchida J, Aoyagi T, Huang WC, Terracina KP, Adams BJ, Rashid OM, Milstien S, Wakai T, Spiegel S, Takabe K. Interstitial Fluid Sphingosine-1-Phosphate in Murine Mammary Gland and Cancer and Human Breast Tissue and Cancer Determined by Novel Methods. J Mammary Gland Biol Neoplasia 2016; 21:9-17. [PMID: 27194029 PMCID: PMC4947521 DOI: 10.1007/s10911-016-9354-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 05/09/2016] [Indexed: 01/15/2023] Open
Abstract
The tumor microenvironment is a determining factor for cancer biology and progression. Sphingosine-1-phosphate (S1P), produced by sphingosine kinases (SphKs), is a bioactive lipid mediator that regulates processes important for cancer progression. Despite its critical roles, the levels of S1P in interstitial fluid (IF), an important component of the tumor microenvironment, have never previously been measured due to a lack of efficient methods for collecting and quantifying IF. The purpose of this study is to clarify the levels of S1P in the IF from murine mammary glands and its tumors utilizing our novel methods. We developed an improved centrifugation method to collect IF. Sphingolipids in IF, blood, and tissue samples were measured by mass spectrometry. In mice with a deletion of SphK1, but not SphK2, levels of S1P in IF from the mammary glands were greatly attenuated. Levels of S1P in IF from mammary tumors were reduced when tumor growth was suppressed by oral administration of FTY720/fingolimod. Importantly, sphingosine, dihydro-sphingosine, and S1P levels, but not dihydro-S1P, were significantly higher in human breast tumor tissue IF than in the normal breast tissue IF. To our knowledge, this is the first reported S1P IF measurement in murine normal mammary glands and mammary tumors, as well as in human patients with breast cancer. S1P tumor IF measurement illuminates new aspects of the role of S1P in the tumor microenvironment.
Collapse
MESH Headings
- Activation, Metabolic
- Animals
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/therapeutic use
- Breast/metabolism
- Breast/pathology
- Breast/surgery
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Cell Line, Tumor
- Extracellular Fluid/drug effects
- Extracellular Fluid/metabolism
- Female
- Fingolimod Hydrochloride/pharmacokinetics
- Fingolimod Hydrochloride/therapeutic use
- Humans
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Lysophospholipids/blood
- Lysophospholipids/metabolism
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice, Inbred BALB C
- Mice, Knockout
- Phosphotransferases (Alcohol Group Acceptor)/genetics
- Phosphotransferases (Alcohol Group Acceptor)/metabolism
- Prodrugs/pharmacokinetics
- Prodrugs/therapeutic use
- Random Allocation
- Sphingosine/analogs & derivatives
- Sphingosine/blood
- Sphingosine/metabolism
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
- Masayuki Nagahashi
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, and Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Broad Street, Richmond, VA, 23298-0011, USA.
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA.
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.
| | - Akimitsu Yamada
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, and Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Broad Street, Richmond, VA, 23298-0011, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Hiroshi Miyazaki
- Section of General Internal Medicine, Kojin Hospital, 1-710 Shikenya, Moriyama, Nagoya, 463-8530, Japan
| | - Jeremy C Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Junko Tsuchida
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Tomoyoshi Aoyagi
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, and Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Broad Street, Richmond, VA, 23298-0011, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Wei-Ching Huang
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, and Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Broad Street, Richmond, VA, 23298-0011, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Krista P Terracina
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, and Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Broad Street, Richmond, VA, 23298-0011, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Barbara J Adams
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, and Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Broad Street, Richmond, VA, 23298-0011, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Omar M Rashid
- Holy Cross Hospital Michael and Dianne Bienes Comprehensive Cancer Center, 4725 North Federal Highway, Fort Lauderdale, FL, 33308, USA
- Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA
- University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, 33136, USA
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA
| | - Kazuaki Takabe
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, and Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Broad Street, Richmond, VA, 23298-0011, USA.
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, and the Massey Cancer Center, PO Box 980011, West Hospital 7-402, 1200 East Marshall Street, Richmond, VA, 23298, USA.
- Breast Surgery, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
30
|
Jin L, Liu WR, Tian MX, Fan J, Shi YH. The SphKs/S1P/S1PR1 axis in immunity and cancer: more ore to be mined. World J Surg Oncol 2016; 14:131. [PMID: 27129720 PMCID: PMC4850705 DOI: 10.1186/s12957-016-0884-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/21/2016] [Indexed: 12/23/2022] Open
Abstract
Over the past two decades, huge amounts of research were launched to understand the functions of sphingosine. Many pathways were uncovered that convey the relative functions of biomacromolecules. In this review, we discuss the recent advances of the role of the SphKs/S1P/S1PR1 axis in immunity and cancer. Finally, we investigate the therapeutic potential of new drugs that target S1P signaling in cancer therapy.
Collapse
Affiliation(s)
- Lei Jin
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 FengLin Road, Shanghai, 200032, China
| | - Wei-Ren Liu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 FengLin Road, Shanghai, 200032, China
| | - Meng-Xin Tian
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 FengLin Road, Shanghai, 200032, China
| | - Jia Fan
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 FengLin Road, Shanghai, 200032, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Ying-Hong Shi
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 FengLin Road, Shanghai, 200032, China.
| |
Collapse
|
31
|
Dimasi DP, Pitson SM, Bonder CS. Examining the Role of Sphingosine Kinase-2 in the Regulation of Endothelial Cell Barrier Integrity. Microcirculation 2016; 23:248-65. [DOI: 10.1111/micc.12271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/25/2016] [Indexed: 12/30/2022]
Affiliation(s)
- David P. Dimasi
- Centre for Cancer Biology; University of South Australia and SA Pathology; Adelaide South Australia Australia
| | - Stuart M. Pitson
- Centre for Cancer Biology; University of South Australia and SA Pathology; Adelaide South Australia Australia
- School of Medicine; University of Adelaide; Adelaide South Australia Australia
- School of Biological Sciences; University of Adelaide; Adelaide South Australia Australia
| | - Claudine S. Bonder
- Centre for Cancer Biology; University of South Australia and SA Pathology; Adelaide South Australia Australia
- School of Medicine; University of Adelaide; Adelaide South Australia Australia
- School of Biological Sciences; University of Adelaide; Adelaide South Australia Australia
| |
Collapse
|
32
|
Sun WY, Dimasi DP, Pitman MR, Zhuang Y, Heddle R, Pitson SM, Grimbaldeston MA, Bonder CS. Topical Application of Fingolimod Perturbs Cutaneous Inflammation. THE JOURNAL OF IMMUNOLOGY 2016; 196:3854-64. [PMID: 27001955 DOI: 10.4049/jimmunol.1501510] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 02/23/2016] [Indexed: 01/13/2023]
Abstract
The prevalence of allergies, including rhinitis, eczema, and anaphylaxis, is rising dramatically worldwide. This increase is especially problematic in children who bear the greatest burden of this rising trend. Increasing evidence identifies neutrophils as primary perpetrators of the more severe and difficult to manage forms of inflammation. A newly recognized mechanism by which neutrophils are recruited during the early phase of histamine-induced inflammation involves the sphingosine kinase (SK)/sphingosine-1-phosphate axis. This study examines whether topical application of fingolimod, an established SK/sphingosine-1-phosphate antagonist already in clinical use to treat multiple sclerosis, may be repurposed to treat cutaneous inflammation. Using two mouse models of ear skin inflammation (histamine- and IgE-mediated passive cutaneous anaphylaxis) we topically applied fingolimod prophylactically, as well as after establishment of the inflammatory response, and examined ear swelling, SK activity, vascular permeability, leukocyte recruitment, and production of proinflammatory mediators. The present study reveals that when applied topically, fingolimod attenuates both immediate and late-phase responses to histamine with reduced extravasation of fluid, SK-1 activity, proinflammatory cytokine and chemokine production, and neutrophil influx and prevents ear swelling. Intravital microscopy demonstrates that histamine-induced neutrophil rolling and adhesion to the postcapillary venules in the mouse ears is significantly attenuated even after 24 h. More importantly, these effects are achievable even once inflammation is established. Translation into humans was also accomplished with epicutaneous application of fingolimod resolving histamine-induced and allergen-induced inflammatory reactions in forearm skin. Overall, this study demonstrates, to our knowledge for the first time, that fingolimod may be repurposed to treat cutaneous inflammation.
Collapse
Affiliation(s)
- Wai Y Sun
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5000, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - David P Dimasi
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5000, Australia
| | - Melissa R Pitman
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5000, Australia
| | - YiZhong Zhuang
- School of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Robert Heddle
- School of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia; Clinical Immunology Unit, Royal Adelaide Hospital, Adelaide, South Australia 5000, Australia; and Discipline of Pediatrics and Child Health, Flinders University, Adelaide, South Australia 5042, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5000, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Michele A Grimbaldeston
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5000, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5000, Australia; School of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia;
| |
Collapse
|
33
|
Frej C, Linder A, Happonen KE, Taylor FB, Lupu F, Dahlbäck B. Sphingosine 1-phosphate and its carrier apolipoprotein M in human sepsis and in Escherichia coli sepsis in baboons. J Cell Mol Med 2016; 20:1170-81. [PMID: 26990127 PMCID: PMC4882985 DOI: 10.1111/jcmm.12831] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/07/2016] [Indexed: 01/01/2023] Open
Abstract
Sphingosine 1‐phosphate (S1P) is an important regulator of vascular integrity and immune cell migration, carried in plasma by high‐density lipoprotein (HDL)‐associated apolipoprotein M (apoM) and by albumin. In sepsis, the protein and lipid composition of HDL changes dramatically. The aim of this study was to evaluate changes in S1P and its carrier protein apoM during sepsis. For this purpose, plasma samples from both human sepsis patients and from an experimental Escherichia coli sepsis model in baboons were used. In the human sepsis cohort, previously studied for apoM, plasma demonstrated disease‐severity correlated decreased S1P levels, the profile mimicking that of plasma apoM. In the baboons, a similar disease‐severity dependent decrease in plasma levels of S1P and apoM was observed. In the lethal E. coli baboon sepsis, S1P decreased already within 6–8 hrs, whereas the apoM decrease was seen later at 12–24 hrs. Gel filtration chromatography of plasma from severe human or baboon sepsis on Superose 6 demonstrated an almost complete loss of S1P and apoM in the HDL fractions. S1P plasma concentrations correlated with the platelet count but not with erythrocytes or white blood cells. The liver mRNA levels of apoM and apoA1 decreased strongly upon sepsis induction and after 12 hr both were almost completely lost. In conclusion, during septic challenge, the plasma levels of S1P drop to very low levels. Moreover, the liver synthesis of apoM decreases severely and the plasma levels of apoM are reduced. Possibly, the decrease in S1P contributes to the decreased endothelial barrier function observed in sepsis.
Collapse
Affiliation(s)
- Cecilia Frej
- Department of Translational Medicine, Division of Clinical Chemistry, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Adam Linder
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, Skåne University Hospital, Lund, Sweden
| | - Kaisa E Happonen
- Department of Translational Medicine, Division of Clinical Chemistry, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Fletcher B Taylor
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Björn Dahlbäck
- Department of Translational Medicine, Division of Clinical Chemistry, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
34
|
Congdon MD, Kharel Y, Brown AM, Lewis SN, Bevan DR, Lynch KR, Santos WL. Structure-Activity Relationship Studies and Molecular Modeling of Naphthalene-Based Sphingosine Kinase 2 Inhibitors. ACS Med Chem Lett 2016; 7:229-34. [PMID: 26985306 DOI: 10.1021/acsmedchemlett.5b00304] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 02/02/2016] [Indexed: 12/26/2022] Open
Abstract
The two isoforms of sphingosine kinase (SphK1 and SphK2) are the only enzymes that phosphorylate sphingosine to sphingosine-1-phosphate (S1P), which is a pleiotropic lipid mediator involved in a broad range of cellular processes including migration, proliferation, and inflammation. SphKs are targets for various diseases such as cancer, fibrosis, and Alzheimer's and sickle cell disease. Herein, we disclose the structure-activity profile of naphthalene-containing SphK inhibitors and molecular modeling studies that reveal a key molecular switch that controls SphK selectivity.
Collapse
Affiliation(s)
- Molly D. Congdon
- Department
of Chemistry, ‡Department of Biochemistry, and §Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Yugesh Kharel
- Department
of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | | | | | | | - Kevin R. Lynch
- Department
of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L. Santos
- Department
of Chemistry, ‡Department of Biochemistry, and §Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
35
|
Graham AC, Temple RM, Obar JJ. Mast cells and influenza a virus: association with allergic responses and beyond. Front Immunol 2015; 6:238. [PMID: 26042121 PMCID: PMC4435071 DOI: 10.3389/fimmu.2015.00238] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/01/2015] [Indexed: 12/07/2022] Open
Abstract
Influenza A virus (IAV) is a widespread infectious agent commonly found in mammalian and avian species. In humans, IAV is a respiratory pathogen that causes seasonal infections associated with significant morbidity in young and elderly populations, and has a large economic impact. Moreover, IAV has the potential to cause both zoonotic spillover infection and global pandemics, which have significantly greater morbidity and mortality across all ages. The pathology associated with these pandemic and spillover infections appear to be the result of an excessive inflammatory response leading to severe lung damage, which likely predisposes the lungs for secondary bacterial infections. The lung is protected from pathogens by alveolar epithelial cells, endothelial cells, tissue resident alveolar macrophages, dendritic cells, and mast cells. The importance of mast cells during bacterial and parasitic infections has been extensively studied; yet, the role of these hematopoietic cells during viral infections is only beginning to emerge. Recently, it has been shown that mast cells can be directly activated in response to IAV, releasing mediators such histamine, proteases, leukotrienes, inflammatory cytokines, and antiviral chemokines, which participate in the excessive inflammatory and pathological response observed during IAV infections. In this review, we will examine the relationship between mast cells and IAV, and discuss the role of mast cells as a potential drug target during highly pathological IAV infections. Finally, we proposed an emerging role for mast cells in other viral infections associated with significant host pathology.
Collapse
Affiliation(s)
- Amy C Graham
- Department of Microbiology and Immunology, Montana State University , Bozeman, MT , USA
| | - Rachel M Temple
- Department of Microbiology and Immunology, Montana State University , Bozeman, MT , USA
| | - Joshua J Obar
- Department of Microbiology and Immunology, Montana State University , Bozeman, MT , USA
| |
Collapse
|
36
|
Draber P, Halova I, Polakovicova I, Kawakami T. Signal transduction and chemotaxis in mast cells. Eur J Pharmacol 2015; 778:11-23. [PMID: 25941081 DOI: 10.1016/j.ejphar.2015.02.057] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 01/08/2023]
Abstract
Mast cells play crucial roles in both innate and adaptive arms of the immune system. Along with basophils, mast cells are essential effector cells for allergic inflammation that causes asthma, allergic rhinitis, food allergy and atopic dermatitis. Mast cells are usually increased in inflammatory sites of allergy and, upon activation, release various chemical, lipid, peptide and protein mediators of allergic reactions. Since antigen/immunoglobulin E (IgE)-mediated activation of these cells is a central event to trigger allergic reactions, innumerable studies have been conducted on how these cells are activated through cross-linking of the high-affinity IgE receptor (FcεRI). Development of mature mast cells from their progenitor cells is under the influence of several growth factors, of which the stem cell factor (SCF) seems to be the most important. Therefore, how SCF induces mast cell development and activation via its receptor, KIT, has been studied extensively, including a cross-talk between KIT and FcεRI signaling pathways. Although our understanding of the signaling mechanisms of the FcεRI and KIT pathways is far from complete, pharmaceutical applications of the knowledge about these pathways are underway. This review will focus on recent progresses in FcεRI and KIT signaling and chemotaxis.
Collapse
Affiliation(s)
- Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, CZ 14220 Prague, Czech Republic.
| | - Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, CZ 14220 Prague, Czech Republic
| | - Iva Polakovicova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, CZ 14220 Prague, Czech Republic
| | - Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle La Jolla, CA 92037, USA; Laboratory for Allergic Disease, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama 230-0045, Japan
| |
Collapse
|
37
|
Kulinski JM, Muñoz-Cano R, Olivera A. Sphingosine-1-phosphate and other lipid mediators generated by mast cells as critical players in allergy and mast cell function. Eur J Pharmacol 2015; 778:56-67. [PMID: 25941085 DOI: 10.1016/j.ejphar.2015.02.058] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 12/20/2022]
Abstract
Sphingosine-1-phosphate (S1P), platelet activating factor (PAF) and eicosanoids are bioactive lipid mediators abundantly produced by antigen-stimulated mast cells that exert their function mostly through specific cell surface receptors. Although it has long been recognized that some of these bioactive lipids are potent regulators of allergic diseases, their exact contributions to disease pathology have been obscured by the complexity of their mode of action and the regulation of their metabolism. Indeed, the effects of such lipids are usually mediated by multiple receptor subtypes that may differ in their signaling mechanisms and functions. In addition, their actions may be elicited by cell surface receptor-independent mechanisms. Furthermore, these lipids may be converted into metabolites that exhibit different functionalities, adding another layer of complexity to their overall biological responses. In some instances, a second wave of lipid mediator synthesis by both mast cell and non-mast cell sources may occur late during inflammation, bringing about additional roles in the altered environment. New evidence also suggests that bioactive lipids in the local environment can fine-tune mast cell maturation and phenotype, and thus their responsiveness. A better understanding of the subtleties of the spatiotemporal regulation of these lipid mediators, their receptors and functions may aid in the pursuit of pharmacological applications for allergy treatments.
Collapse
Affiliation(s)
- Joseph M Kulinski
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Rosa Muñoz-Cano
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Ana Olivera
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
The Granuloma Response Controlling Cryptococcosis in Mice Depends on the Sphingosine Kinase 1-Sphingosine 1-Phosphate Pathway. Infect Immun 2015; 83:2705-13. [PMID: 25895971 DOI: 10.1128/iai.00056-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/13/2015] [Indexed: 12/31/2022] Open
Abstract
Cryptococcus neoformans is a fungal pathogen that causes pulmonary infections, which may progress into life-threatening meningitis. In commonly used mouse models of C. neoformans infections, fungal cells are not contained in the lungs, resulting in dissemination to the brain. We have previously reported the generation of an engineered C. neoformans strain (C. neoformans Δgcs1) which can be contained in lung granulomas in the mouse model and have shown that granuloma formation is dependent upon the enzyme sphingosine kinase 1 (SK1) and its product, sphingosine 1-phosphate (S1P). In this study, we have used four mouse models, CBA/J and C57BL6/J (both immunocompetent), Tgε26 (an isogenic strain of strain CBA/J lacking T and NK cells), and SK(-/-) (an isogenic strain of strain C57BL6/J lacking SK1), to investigate how the granulomatous response and SK1-S1P pathway are interrelated during C. neoformans infections. S1P and monocyte chemotactic protein-1 (MCP-1) levels were significantly elevated in the bronchoalveolar lavage fluid of all mice infected with C. neoformans Δgcs1 but not in mice infected with the C. neoformans wild type. SK1(-/-) mice did not show elevated levels of S1P or MCP-1. Primary neutrophils isolated from SK1(-/-) mice showed impaired antifungal activity that could be restored by the addition of extracellular S1P. In addition, high levels of tumor necrosis factor alpha were found in the mice infected with C. neoformans Δgcs1 in comparison to the levels found in mice infected with the C. neoformans wild type, and their levels were also dependent on the SK1-S1P pathway. Taken together, these results suggest that the SK1-S1P pathway promotes host defense against C. neoformans infections by regulating cytokine levels, promoting extracellular killing by phagocytes, and generating a granulomatous response.
Collapse
|
39
|
Patwardhan NN, Morris EA, Kharel Y, Raje MR, Gao M, Tomsig JL, Lynch KR, Santos WL. Structure-activity relationship studies and in vivo activity of guanidine-based sphingosine kinase inhibitors: discovery of SphK1- and SphK2-selective inhibitors. J Med Chem 2015; 58:1879-1899. [PMID: 25643074 DOI: 10.1021/jm501760d] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a pleiotropic signaling molecule that acts as a ligand for five G-protein coupled receptors (S1P1-5) whose downstream effects are implicated in a variety of important pathologies including sickle cell disease, cancer, inflammation, and fibrosis. The synthesis of S1P is catalyzed by sphingosine kinase (SphK) isoforms 1 and 2, and hence, inhibitors of this phosphorylation step are pivotal in understanding the physiological functions of SphKs. To date, SphK1 and 2 inhibitors with the potency, selectivity, and in vivo stability necessary to determine the potential of these kinases as therapeutic targets are lacking. Herein, we report the design, synthesis, and structure-activity relationship studies of guanidine-based SphK inhibitors bearing an oxadiazole ring in the scaffold. Our studies demonstrate that SLP120701, a SphK2-selective inhibitor (Ki = 1 μM), decreases S1P levels in histiocytic lymphoma (U937) cells. Surprisingly, homologation with a single methylene unit between the oxadiazole and heterocyclic ring afforded a SphK1-selective inhibitor in SLP7111228 (Ki = 48 nM), which also decreased S1P levels in cultured U937 cells. In vivo application of both compounds, however, resulted in contrasting effect in circulating levels of S1P. Administration of SLP7111228 depressed blood S1P levels while SLP120701 increased levels of S1P. Taken together, these compounds provide an in vivo chemical toolkit to interrogate the effect of increasing or decreasing S1P levels and whether such a maneuver can have implications in disease states.
Collapse
Affiliation(s)
- Neeraj N Patwardhan
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Emily A Morris
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Mithun R Raje
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Ming Gao
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Jose L Tomsig
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
40
|
Abstract
The transfer of the gamma phosphate from ATP to sphingosine (Sph) to generate a small signaling molecule, sphingosine 1-phosphate (S1P), is catalyzed by sphingosine kinases (SphK), which exist as two isoforms, SphK1 and SphK2. SphK is a key regulator of S1P and the S1P:Sph/ceramide ratio. Increases in S1P levels have been linked to diseases including sickle cell disease, cancer, and fibrosis. Therefore, SphKs are potential targets for drug discovery. However, the current chemical biology toolkit needed to validate these enzymes as drug targets is inadequate. With this review, we survey in vivo active SphK inhibitors and highlight the need for developing more potent and selective inhibitors.
Collapse
Affiliation(s)
- Webster L. Santos
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Kevin R. Lynch
- Department
of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
41
|
Suzuki R, Scheffel J, Rivera J. New insights on the signaling and function of the high-affinity receptor for IgE. Curr Top Microbiol Immunol 2015; 388:63-90. [PMID: 25553795 DOI: 10.1007/978-3-319-13725-4_4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Clustering of the high-affinity receptor for immunoglobulin E (FcεRI) through the interaction of receptor-bound immunoglobulin E (IgE) antibodies with their cognate antigen is required to couple IgE antibody production to cellular responses and physiological consequences. IgE-induced responses through FcεRI are well known to defend the host against certain infectious agents and to lead to unwanted allergic responses to normally innocuous substances. However, the cellular and/or physiological response of individuals that produce IgE antibodies may be markedly different and such antibodies (even to the same antigenic epitope) can differ in their antigen-binding affinity. How affinity variation in the interaction of FcεRI-bound IgE antibodies with antigen is interpreted into cellular responses and how the local environment may influence these responses is of interest. In this chapter, we focus on recent advances that begin to unravel how FcεRI distinguishes differences in the affinity of IgE-antigen interactions and how such discrimination along with surrounding environmental stimuli can shape the (patho) physiological response.
Collapse
Affiliation(s)
- Ryo Suzuki
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | |
Collapse
|
42
|
The sphingosine-1-phosphate/sphingosine-1-phosphate receptor 2 axis regulates early airway T-cell infiltration in murine mast cell-dependent acute allergic responses. J Allergy Clin Immunol 2014; 135:1008-1018.e1. [PMID: 25512083 DOI: 10.1016/j.jaci.2014.10.044] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 10/28/2014] [Accepted: 10/31/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid produced by mast cells (MCs) on cross-linking of their high-affinity receptors for IgE by antigen that can amplify MC responses by binding to its S1P receptors. An acute MC-dependent allergic reaction can lead to systemic shock, but the early events of its development in lung tissues have not been investigated, and S1P functions in the onset of allergic processes remain to be examined. OBJECTIVE We used a highly specific neutralizing anti-S1P antibody (mAb) and the sphingosine-1-phosphate receptor 2 (S1PR2) antagonist JTE-013 to study the signaling contributions of S1P and S1PR2 to MC- and IgE-dependent airway allergic responses in mice within minutes after antigen challenge. METHODS Allergic reaction was triggered by a single intraperitoneal dose of antigen in sensitized mice pretreated intraperitoneally with anti-S1P, isotype control mAb, JTE-013, or vehicle before antigen challenge. RESULTS Kinetics experiments revealed early pulmonary infiltration of mostly T cells around blood vessels of sensitized mice 20 minutes after antigen exposure. Pretreatment with anti-S1P mAb inhibited in vitro MC activation, as well as in vivo development of airway infiltration and MC activation, reducing serum levels of histamine, cytokines, and the chemokines monocyte chemoattractant protein 1/CCL2, macrophage inflammatory protein 1α/CCL3, and RANTES/CCL5. S1PR2 antagonism or deficiency or MC deficiency recapitulated these results. Both in vitro and in vivo experiments demonstrated MC S1PR2 dependency for chemokine release and the necessity for signal transducer and activator of transcription 3 activation. CONCLUSION Activation of S1PR2 by S1P and downstream signal transducer and activator of transcription 3 signaling in MCs regulate early T-cell recruitment to antigen-challenged lungs through chemokine production.
Collapse
|
43
|
Rosenstein RK, Bezbradica JS, Yu S, Medzhitov R. Signaling pathways activated by a protease allergen in basophils. Proc Natl Acad Sci U S A 2014; 111:E4963-71. [PMID: 25369937 PMCID: PMC4246326 DOI: 10.1073/pnas.1418959111] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Allergic diseases represent a significant burden in industrialized countries, but why and how the immune system responds to allergens remain largely unknown. Because many clinically significant allergens have proteolytic activity, and many helminths express proteases that are necessary for their life cycles, host mechanisms likely have evolved to detect the proteolytic activity of helminth proteases, which may be incidentally activated by protease allergens. A cysteine protease, papain, is a prototypic protease allergen that can directly activate basophils and mast cells, leading to the production of cytokines, including IL-4, characteristic of the type 2 immune response. The mechanism of papain's immunogenic activity remains unknown. Here we have characterized the cellular response activated by papain in basophils. We find that papain-induced IL-4 production requires calcium flux and activation of PI3K and nuclear factor of activated T cells. Interestingly, papain-induced IL-4 production was dependent on the immunoreceptor tyrosine-based activation motif (ITAM) adaptor protein Fc receptor γ-chain, even though the canonical ITAM signaling was not activated by papain. Collectively, these data characterize the downstream signaling pathway activated by a protease allergen in basophils.
Collapse
Affiliation(s)
- Rachel K Rosenstein
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Jelena S Bezbradica
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Shuang Yu
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Ruslan Medzhitov
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
44
|
Borriello F, Granata F, Varricchi G, Genovese A, Triggiani M, Marone G. Immunopharmacological modulation of mast cells. Curr Opin Pharmacol 2014; 17:45-57. [PMID: 25063971 DOI: 10.1016/j.coph.2014.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 07/02/2014] [Accepted: 07/04/2014] [Indexed: 12/31/2022]
Abstract
Mast cells produce a wide spectrum of mediators and they have been implicated in several physiopathological conditions (e.g. allergic reactions and certain tumors). Pharmacologic agents that modulate the release of mediators from mast cells has helped to elucidate the biochemical mechanisms by which immunological and non-immunological stimuli activate these cells. Furthermore, the study of surface receptors and signaling pathways associated with mast cell activation revealed novel pharmacologic targets. Thus, the development of pharmacologic agents based on this new wave of knowledge holds promise for the treatment of several mast cell-mediated disorders.
Collapse
Affiliation(s)
- Francesco Borriello
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, Via Pansini 5, 80131 Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, Via Pansini 5, 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, Via Pansini 5, 80131 Naples, Italy
| | - Arturo Genovese
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, Via Pansini 5, 80131 Naples, Italy
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, School of Medicine, Salerno, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
45
|
Kim EY, Sturgill JL, Hait NC, Avni D, Valencia EC, Maceyka M, Lima S, Allegood J, Huang WC, Zhang S, Milstien S, Conrad D, Spiegel S. Role of sphingosine kinase 1 and sphingosine-1-phosphate in CD40 signaling and IgE class switching. FASEB J 2014; 28:4347-58. [PMID: 25002116 DOI: 10.1096/fj.14-251611] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The tumor necrosis factor (TNF) receptor family member CD40 plays an essential role in the activation of antigen-presenting cells, B cell maturation, and immunoglobulin (Ig) class switching critical for adaptive immunity. Although the bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) and the kinase that produces it, sphingosine kinase 1 (SphK1), have long been implicated in the actions of TNF mediated by engagement of TNFR1, nothing is yet known of their role in CD40-mediated events. We have now found that ligation of CD40 activates and translocates SphK1 to the plasma membrane, leading to generation of S1P. SphK1 inhibition in human tonsil B cells, as well as inhibition or deletion of SphK1 in mouse splenic B cells, significantly reduced CD40-mediated Ig class switching and plasma cell differentiation ex vivo. Optimal activation of downstream CD40 signaling pathways, including NF-κB, p38, and JNK, also required SphK1. In mice treated with a SphK1 inhibitor or in SphK1(-/-) mice, isotype switching to antigen-specific IgE was decreased in vivo by 70 and 55%, respectively. Our results indicate that SphK1 is important for CD40-mediated B cell activation and regulation of humoral responses and suggest that targeting SphK1 might be a useful therapeutic approach to control antigen-specific IgE production.
Collapse
Affiliation(s)
- Eugene Y Kim
- Department of Biochemistry and Molecular Biology
| | | | - Nitai C Hait
- Department of Biochemistry and Molecular Biology
| | - Dorit Avni
- Department of Biochemistry and Molecular Biology
| | | | | | | | | | | | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | | | | | | |
Collapse
|
46
|
Yan W, Zhang F, Zhang R, Zhang X, Wang Y, Zhou F, Xia Y, Liu P, Gao C, Wang H, Zhang L, Zhou J, Gao F, Gao E, Koch WJ, Wang H, Cheng H, Qu Y, Tao L. Adiponectin regulates SR Ca(2+) cycling following ischemia/reperfusion via sphingosine 1-phosphate-CaMKII signaling in mice. J Mol Cell Cardiol 2014; 74:183-92. [PMID: 24852843 DOI: 10.1016/j.yjmcc.2014.05.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/15/2014] [Accepted: 05/08/2014] [Indexed: 01/01/2023]
Abstract
The adipocyte-secreted hormone adiponectin (APN) exerts protective effects on the heart under stress conditions. Recent studies have demonstrated that APN induces a marked Ca(2+) influx in skeletal muscle. However, whether APN modulates [Ca(2+)]i activity, especially [Ca(2+)]i transients in cardiomyocytes, is still unknown. This study was designed to determine whether APN modulates [Ca(2+)]i transients in cardiomyocytes. Adult male wild-type (WT) and APN knockout (APN KO) mice were subjected to myocardial ischemia/reperfusion (I/R, 30min/30min) injury. CaMKII-PLB phosphorylation and SR Ca(2+)-ATPase (SERCA2) activity were downregulated in I/R hearts of WT mice and further decreased in those of APN KO mice. Both the globular domain of APN and full-length APN significantly reversed the decrease in CaMKII-PLB phosphorylation and SERCA2 activity in WT and APN KO mice. Interestingly, compared with WT littermates, single myocytes isolated from APN KO mice had remarkably decreased [Ca(2+)]i transients, cell shortening, and a prolonged Ca(2+) decay rate. Further examination revealed that APN enhances SERCA2 activity via CaMKII-PLB signaling. In in vivo and in vitro experiments, both APN receptor 1/2 and S1P were necessary for the APN-stimulated CaMKII-PLB-SERCA2 activation. In addition, S1P activated CaMKII-PLB signaling in neonatal cardiomyocytes in a dose dependent manner and improved [Ca(2+)]i transients in APN KO myocytes via the S1P receptor (S1PR1/3). Further in vivo experiments revealed that pharmacological inhibition of S1PR1/3 and SERCA2 siRNA suppressed APN-mediated cardioprotection during I/R. These data demonstrate that S1P is a novel regulator of SERCA2 that activates CaMKII-PLB signaling and mediates APN-induced cardioprotection.
Collapse
Affiliation(s)
- Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ronghuai Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing Zhang
- Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Yanru Wang
- Institutes of Molecular Medicine, Peking University, Beijing 100083, China
| | - Fen Zhou
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Peilin Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Han Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lijian Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jingjun Zhou
- Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Feng Gao
- Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an 710032, China
| | - Erhe Gao
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA 19107, USA
| | - Walter J Koch
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA 19107, USA
| | - Haichang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Heping Cheng
- Institutes of Molecular Medicine, Peking University, Beijing 100083, China
| | - Yan Qu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
47
|
Yip KH, Kolesnikoff N, Yu C, Hauschild N, Taing H, Biggs L, Goltzman D, Gregory PA, Anderson PH, Samuel MS, Galli SJ, Lopez AF, Grimbaldeston MA. Mechanisms of vitamin D₃ metabolite repression of IgE-dependent mast cell activation. J Allergy Clin Immunol 2014; 133:1356-64, 1364.e1-14. [PMID: 24461581 PMCID: PMC4154631 DOI: 10.1016/j.jaci.2013.11.030] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 10/15/2013] [Accepted: 11/15/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND Mast cells have gained notoriety based on their detrimental contributions to IgE-mediated allergic disorders. Although mast cells express the vitamin D receptor (VDR), it is not clear to what extent 1α,25-dihydroxyvitamin D3 (1α,25[OH]2D3) or its predominant inactive precursor metabolite in the circulation, 25-hydroxyvitamin D3 (25OHD3), can influence IgE-mediated mast cell activation and passive cutaneous anaphylaxis (PCA) in vivo. OBJECTIVE We sought to assess whether the vitamin D3 metabolites 25OHD3 and 1α,25(OH)2D3 can repress IgE-dependent mast cell activation through mast cell-25-hydroxyvitamin D-1α-hydroxylase (CYP27B1) and mast cell-VDR activity. METHODS We measured the extent of vitamin D3 suppression of IgE-mediated mast cell degranulation and mediator production in vitro, as well as the vitamin D3-induced curtailment of PCA responses in WBB6F1-Kit(W/W-v) or C57BL/6J-Kit(W-sh/W-sh) mice engrafted with mast cells that did or did not express VDR or CYP27B1. RESULTS Here we show that mouse and human mast cells can convert 25OHD3 to 1α,25(OH)2D3 through CYP27B1 activity and that both of these vitamin D3 metabolites suppressed IgE-induced mast cell-derived proinflammatory and vasodilatory mediator production in a VDR-dependent manner in vitro. Furthermore, epicutaneously applied vitamin D3 metabolites significantly reduced the magnitude of skin swelling associated with IgE-mediated PCA reactions in vivo; a response that required functional mast cell-VDRs and mast cell-CYP27B1. CONCLUSION Taken together, our findings provide a mechanistic explanation for the anti-inflammatory effects of vitamin D3 on mast cell function by demonstrating that mast cells can actively metabolize 25OHD3 to dampen IgE-mediated mast cell activation in vitro and in vivo.
Collapse
Affiliation(s)
- Kwok-Ho Yip
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Natasha Kolesnikoff
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Chunping Yu
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia; University of Adelaide, Adelaide, Australia
| | - Nicholas Hauschild
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Houng Taing
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia; University of Adelaide, Adelaide, Australia
| | - Lisa Biggs
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - David Goltzman
- Departments of Medicine and Physiology, McGill University, Montreal, Quebec, Canada
| | - Philip A Gregory
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | | | - Michael S Samuel
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia; University of Adelaide, Adelaide, Australia; University of South Australia, Adelaide, Australia
| | - Stephen J Galli
- Departments of Pathology and of Microbiology and Immunology, Stanford University School of Medicine, Stanford, Calif
| | - Angel F Lopez
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia; University of Adelaide, Adelaide, Australia; University of South Australia, Adelaide, Australia
| | - Michele A Grimbaldeston
- Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia; University of Adelaide, Adelaide, Australia; University of South Australia, Adelaide, Australia.
| |
Collapse
|
48
|
Sakuma M, Shirai Y, Ueyama T, Saito N. Diacylglycerol kinase γ regulates antigen-induced mast cell degranulation by mediating Ca(2+) influxes. Biochem Biophys Res Commun 2014; 445:340-5. [PMID: 24513282 DOI: 10.1016/j.bbrc.2014.01.197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 01/30/2014] [Indexed: 10/25/2022]
Abstract
Diacylglycerol (DAG) is an important lipid that acts as a signaling messenger during mast cell degranulation after allergen cross-linking of immunoglobulin (Ig) E-bound FcεRI receptors. In this study, we determined the role of diacylglycerol kinase (DGK), which negatively regulates DAG-dependent signaling by converting DAG to phosphatidic acid (PA), in the regulation of mast cell degranulation. Treating RBL (rat basophilic leukemia)-2H3 mast cells with a type I DGK inhibitor significantly reduced antigen-induced degranulation and PA production. Among type I DGK isoforms, we observed that DGKα and DGKγ mRNAs were expressed in RBL-2H3 mast cells using reverse transcription polymerase chain reaction. DGKγ knockdown, but not DGKα, by isoform-specific short hairpin RNAs reduced mast cell degranulation and Ca(2+) influxes from the extracellular environment. These results suggest that DGKγ regulates mast cell degranulation after FcεRI cross-linking through mobilization of intracellular Ca(2+) through Ca(2+) influxes.
Collapse
Affiliation(s)
- Megumi Sakuma
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| | - Yasuhito Shirai
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| | - Naoaki Saito
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
49
|
Reitsema V, Bouma H, Willem Kok J. Sphingosine-1-phosphate transport and its role in immunology. AIMS MOLECULAR SCIENCE 2014. [DOI: 10.3934/molsci.2014.4.183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
50
|
Abstract
Sphingosine 1-phosphate (S1P), a lipid mediator produced by sphingolipid metabolism, promotes endothelial cell spreading, vascular maturation/stabilization, and barrier function. S1P is present at high concentrations in the circulatory system, whereas in tissues its levels are low. This so-called vascular S1P gradient is essential for S1P to regulate much physiological and pathophysiological progress such as the modulation of vascular permeability. Cellular sources of S1P in blood has only recently begun to be identified. In this review, we summarize the current understanding of S1P in regulating vascular integrity. In particular, we discuss the recent discovery of the endothelium-protective functions of HDL-bound S1P which is chaperoned by apolipoprotein M.
Collapse
|