1
|
Lee MK, Jeong HH, Kim MJ, Seo JS, Hwang JY, Jung WK, Moon KM, Lee I, Lee B. The Beneficial Roles of Sargassum spp. in Skin Disorders. J Med Food 2024; 27:359-368. [PMID: 38526569 DOI: 10.1089/jmf.2023.k.0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024] Open
Abstract
As the body's largest organ, the skin is located at the internal and external environment interface, serving as a line of defense against various harmful stressors. Recently, marine-derived physiologically active ingredients have attracted considerable attention in the cosmeceutical industry due to their beneficial effects on skin health. Sargassum, a genus of brown macroalgae, has traditionally been consumed as food and medicine in several countries and is rich in bioactive compounds such as meroterpenoids, sulfated polysaccharides, fucoidan, fucoxanthin, flavonoids, and terpenoids. Sargassum spp. have various beneficial effects on skin disorders. They help with atopic dermatitis by improving skin barrier protection and reducing inflammation. Several species show potential in treating acne by inhibiting bacterial growth and reducing inflammation. Some species, such as Sargassum horneri, demonstrate antiallergic effects by modulating mast cell activity. Certain Sargassum species exhibit anticancer activity by inhibiting tumor growth and promoting apoptosis, and some species help with wound healing by promoting angiogenesis and reducing oxidative stress. Overall, Sargassum spp. demonstrate potential for treating and managing various skin conditions. Therefore, the bioactive compounds of Sargassum spp. may be natural ingredients with a wide range of functional properties for preventing and treating skin disorders. The present review focused on the various biological effects of Sargassum extracts and derived compounds on skin disorders.
Collapse
Affiliation(s)
- Min-Kyeong Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, Korea
- Biotechnology Research Division, National Institute of Fisheries Science, Busan, Republic of Korea
| | - Hyeon Hak Jeong
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Korea
| | - Myeong-Jin Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan, Korea
| | - Jae Seong Seo
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Korea
| | - Ji Young Hwang
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Korea
| | - Won-Kyo Jung
- Division of Biomedical Engineering and Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Korea
| | - Kyoung Mi Moon
- Department of Food Science and Nutrition, Pukyong National University, Busan, Korea
| | - Incheol Lee
- Department of Ocean Engineering, Pukyong National University, Busan, Korea
| | - Bonggi Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, Korea
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Korea
| |
Collapse
|
2
|
Schulman ES, Nishi H, Pelleg A. Degranulation of human mast cells: modulation by P2 receptors' agonists. Front Immunol 2023; 14:1216580. [PMID: 37868982 PMCID: PMC10585249 DOI: 10.3389/fimmu.2023.1216580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/04/2023] [Indexed: 10/24/2023] Open
Abstract
Since the late 1970s, there has been an alarming increase in the incidence of asthma and its morbidity and mortality. Acute obstruction and inflammation of allergic asthmatic airways are frequently caused by inhalation of exogenous substances such as allergens cross-linking IgE receptors expressed on the surface of the human lung mast cells (HLMC). The degree of constriction of human airways produced by identical amounts of inhaled allergens may vary from day to day and even hour to hour. Endogenous factors in the human mast cell (HMC)'s microenvironment during allergen exposure may markedly modulate the degranulation response. An increase in allergic responsiveness may significantly enhance bronchoconstriction and breathlessness. This review focuses on the role that the ubiquitous endogenous purine nucleotide, extracellular adenosine 5'-triphosphate (ATP), which is a component of the damage-associated molecular patterns, plays in mast cells' physiology. ATP activates P2 purinergic cell-surface receptors (P2R) to trigger signaling cascades resulting in heightened inflammatory responses. ATP is the most potent enhancer of IgE-mediated HLMC degranulation described to date. Current knowledge of ATP as it relates to targeted receptor(s) on HMC along with most recent studies exploring HMC post-receptor activation pathways are discussed. In addition, the reviewed studies may explain why brief, minimal exposures to allergens (e.g., dust, cat, mouse, and grass) can unpredictably lead to intense clinical reactions. Furthermore, potential therapeutic approaches targeting ATP-related enhancement of allergic reactions are presented.
Collapse
Affiliation(s)
- Edward S. Schulman
- Division of Pulmonary, Critical Care and Allergy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Haruhisa Nishi
- Department of Pharmacology, Jikei University School of Medicine, Tokyo, Japan
| | - Amir Pelleg
- Danmir Therapeutics, LLC, Haverford, PA, United States
| |
Collapse
|
3
|
Kotani Y, Sumiyoshi M, Sasada M, Watanabe T, Matsuda S. Arf1 facilitates mast cell proliferation via the mTORC1 pathway. Sci Rep 2022; 12:22297. [PMID: 36566324 PMCID: PMC9789986 DOI: 10.1038/s41598-022-26925-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Mast cells are one of major players in allergic responses. Mast cell activation via the high affinity IgE receptor (FcεRI) causes degranulation and release of de novo synthesized proinflammatory cytokines in a process that involves vesicle trafficking. Considering that the GTPase ADP-ribosylation factor 1 (Arf1) orchestrates and maintains membrane traffic and organelle structure, it seems likely that Arf1 contributes to mast cell activation. Actually, it has been reported that pharmaceutical blockade of the Arf1 pathway suppresses cytokine secretion and mast cell degranulation. However, physiological roles of Arf1 in mast cells remain elusive. Here, by using a genetic approach, we demonstrate that Arf1 is required for optimal mTORC1 activation upon IL-3 and facilitates mast cell proliferation. On the other hand, contrary to our expectation, Arf1-deficiency had little impact on FcεRI-induced degranulation nor cytokine secretion. Our findings reveal an unexpected role of Arf1 in mast cell expansion and its potential as a therapeutic target in the mast cell proliferative disorders.
Collapse
Affiliation(s)
- Yui Kotani
- grid.410783.90000 0001 2172 5041Department of Cell Signaling, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010 Japan ,grid.174568.90000 0001 0059 3836Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women’s University, Nara, 630-8506 Japan
| | - Mami Sumiyoshi
- grid.410783.90000 0001 2172 5041Department of Cell Signaling, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010 Japan
| | - Megumi Sasada
- grid.174568.90000 0001 0059 3836Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women’s University, Nara, 630-8506 Japan
| | - Toshio Watanabe
- grid.174568.90000 0001 0059 3836Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women’s University, Nara, 630-8506 Japan
| | - Satoshi Matsuda
- grid.410783.90000 0001 2172 5041Department of Cell Signaling, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010 Japan
| |
Collapse
|
4
|
Niu C, Li M, Chen Y, Zhang X, Zhu S, Zhou X, Zhou L, Li Z, Xu J, Hu JF, Wang Y, Cui J. LncRNA NCAL1 potentiates natural killer cell cytotoxicity through the Gab2-PI3K-AKT pathway. Front Immunol 2022; 13:970195. [PMID: 36248894 PMCID: PMC9554105 DOI: 10.3389/fimmu.2022.970195] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/31/2022] [Indexed: 11/24/2022] Open
Abstract
Natural killer (NK) cells perform immune surveillance functions in tumors. The antitumor effects of NK cells are closely related to tumor occurrence and development. However, the molecular factors that determine NK cell antitumor activity remain to be characterized. In the present study, we identified a novel long noncoding RNA (lncRNA), NK cell activity-associated lncRNA 1 (NCAL1), and investigated its function in NK cells. NCAL1 was primarily located in NK cell nuclei, where it functioned by activating Gab2, a scaffold protein with an essential role in immune cells. Gab2 positively regulated the killing activity of NK cells. Mechanistically, NCAL1 upregulated Gab2 epigenetically by binding to the Gab2 promoter, which decreased methylation, recruited the transcription factor Sp1, and increased H3K4me3 and H3K27ac levels in the Gab2 promoter. Furthermore, NCAL1 enhanced the cytotoxicity of NK cells toward tumor cells through the Gab2-PI3K-AKT pathway. Thus, NCAL1 potentiates NK cell cytotoxicity and is a promising therapeutic target to improve NK cell therapy.
Collapse
Affiliation(s)
- Chao Niu
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Min Li
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Yongchong Chen
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Xiaoying Zhang
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Shan Zhu
- Department of Translational Medicine, First Hospital, Jilin University, Changchun, China
| | - Xin Zhou
- Cancer Institute, First Hospital, Jilin University, Changchun, China
| | - Lei Zhou
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Zhaozhi Li
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Jianting Xu
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Ji-fan Hu
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
- Stanford University Medical School, Veterans Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
- *Correspondence: Jiuwei Cui, ; Yufeng Wang, ; Ji-fan Hu, ;
| | - Yufeng Wang
- Cancer Institute, First Hospital, Jilin University, Changchun, China
- *Correspondence: Jiuwei Cui, ; Yufeng Wang, ; Ji-fan Hu, ;
| | - Jiuwei Cui
- Department of Cancer Center, First Hospital, Jilin University, Changchun, China
- *Correspondence: Jiuwei Cui, ; Yufeng Wang, ; Ji-fan Hu, ;
| |
Collapse
|
5
|
Burchett JR, Dailey JM, Kee SA, Pryor DT, Kotha A, Kankaria RA, Straus DB, Ryan JJ. Targeting Mast Cells in Allergic Disease: Current Therapies and Drug Repurposing. Cells 2022; 11:3031. [PMID: 36230993 PMCID: PMC9564111 DOI: 10.3390/cells11193031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
The incidence of allergic disease has grown tremendously in the past three generations. While current treatments are effective for some, there is considerable unmet need. Mast cells are critical effectors of allergic inflammation. Their secreted mediators and the receptors for these mediators have long been the target of allergy therapy. Recent drugs have moved a step earlier in mast cell activation, blocking IgE, IL-4, and IL-13 interactions with their receptors. In this review, we summarize the latest therapies targeting mast cells as well as new drugs in clinical trials. In addition, we offer support for repurposing FDA-approved drugs to target mast cells in new ways. With a multitude of highly selective drugs available for cancer, autoimmunity, and metabolic disorders, drug repurposing offers optimism for the future of allergy therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John J. Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
6
|
Longé C, Bratti M, Kurowska M, Vibhushan S, David P, Desmeure V, Huang JD, Fischer A, de Saint Basile G, Sepulveda FE, Blank U, Ménasché G. Rab44 regulates murine mast cell-driven anaphylaxis through kinesin-1-dependent secretory granule translocation. J Allergy Clin Immunol 2022; 150:676-689. [PMID: 35469841 DOI: 10.1016/j.jaci.2022.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/15/2022] [Accepted: 04/08/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Mast cells (MCs) are key effectors of the allergic response. Following the cross-linking of IgE receptors (FcεRIs), they release crucial inflammatory mediators through degranulation. Although degranulation depends critically on secretory granule (SG) trafficking towards the plasma membrane, the molecular machinery underlying this transport has not been fully characterized. OBJECTIVE Here, we analyzed the function of Rab44, a large atypical Rab GTPase highly expressed in MC, in MC degranulation process. METHODS Murine KO mouse models (KORab44 and DKOKif5b/Rab44) were used to perform passive cutaneous anaphylaxis (PCA) experiments and analyze granule translocation in derived bone-marrow-derived MCs (BMMCs) during degranulation. RESULTS We demonstrate that mice lacking Rab44 (KORab44) in their BMMCs are impaired in their ability to translocate and degranulate SGs at the plasma membrane upon FcεRI stimulation. Accordingly, KORab44 mice were less sensitive to IgE-mediated passive cutaneous anaphylaxis in vivo. A lack of Rab44 did not impair early FcεRI-stimulated signaling pathways, microtubule reorganization, lipid mediator or cytokine secretion. Mechanistically, Rab44 appears to interact with and function as part of the previously described kinesin-1-dependent transport pathway. CONCLUSIONS Our results highlight a novel role of Rab44 as a regulator of SG transport during degranulation and anaphylaxis acting through the kinesin-1-dependent microtubule transport machinery. Rab44 can thus be considered as a potential target for modulating MC degranulation and inhibiting IgE-mediated allergic reactions.
Collapse
Affiliation(s)
- Cyril Longé
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015 Paris France
| | - Manuela Bratti
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, Paris, France; Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Mathieu Kurowska
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015 Paris France
| | - Shamila Vibhushan
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, Paris, France; Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Pierre David
- Transgenesis Facility, Laboratoire d'Expérimentation Animale et Transgénèse (LEAT), Imagine Institute, Structure Fédérative de Recherche Necker INSERM US24/CNRS UMS3633, F-75015, Paris, France
| | - Valère Desmeure
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015 Paris France
| | - Jian-Dong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Alain Fischer
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015 Paris France; Immunology and Pediatric Hematology Department, Necker Children's Hospital, AP-HP, F-75015 Paris, France; Collège de France, F-75005 Paris, France
| | - Geneviève de Saint Basile
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015 Paris France; Centre d'Etude des Déficits Immunitaires, AP-HP, Hôpital Necker-Enfants Malades, F-75015, Paris, France
| | - Fernando E Sepulveda
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015 Paris France; Centre National de la Recherche Scientifique, F-75015, Paris. France
| | - Ulrich Blank
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, Paris, France; Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Gaël Ménasché
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015 Paris France
| |
Collapse
|
7
|
Jang HY, Ha DH, Rah SY, Lee DH, Lee SM, Park BH. Sirtuin 6 is a negative regulator of FcεRI signaling and anaphylactic responses. J Allergy Clin Immunol 2022; 149:156-167.e7. [PMID: 34051221 DOI: 10.1016/j.jaci.2021.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Binding IgE to a cognate allergen causes aggregation of Fcε receptor I (FcεRI) in mast cells, resulting in activation of receptor-associated Src family tyrosine kinases, including Lyn and Syk. Protein tyrosine phosphatase, receptor type C (PTPRC), also known as CD45, has emerged as a positive regulator of FcεRI signaling by dephosphorylation of the inhibitory tyrosine of Lyn. OBJECTIVE Sirtuin 6 (Sirt6), a NAD+-dependent deacetylase, exhibits an anti-inflammatory property. It remains to be determined, however, whether Sirt6 attenuates mast cell-associated diseases, including anaphylaxis. METHODS FcεRI signaling and mast cell degranulation were measured after IgE cross-linking in murine bone marrow-derived mast cells (BMMCs) and human cord blood-derived mast cells. To investigate the function of Sirt6 in mast cell activation in vivo, we used mast cell-dependent animal models of passive systemic anaphylaxis (PSA) and passive cutaneous anaphylaxis (PCA). RESULTS Sirt6-deficient BMMCs augmented IgE-FcεRI-mediated signaling and degranulation compared to wild-type BMMCs. Reconstitution of mast cell-deficient KitW-sh/W-sh mice with BMMCs received from Sirt6 knockout mice developed more severe PSA and PCA compared to mice engrafted with wild-type BMMCs. Similarly, genetic overexpression or pharmacologic activation of Sirt6 suppressed mast cell degranulation and blunted responses to PCA. Mechanistically, Sirt6 deficiency increased PTPRC transcription via acetylating histone H3, leading to enhanced aggregation of FcεRI in BMMCs. Finally, we recapitulated the Sirt6 regulation of PTPRC and FcεRI signaling in human mast cells. CONCLUSIONS Sirt6 acts as a negative regulator of FcεRI signaling cascade in mast cells by suppressing PTPRC transcription. Activation of Sirt6 may therefore represent a promising and novel therapeutic strategy for anaphylaxis.
Collapse
Affiliation(s)
- Hyun-Young Jang
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Korea
| | - Do Hyun Ha
- Division of Biotechnology, College of Environmental and Bioresource Sciences, Chonbuk National University, Iksan, Korea
| | - So-Young Rah
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Korea
| | - Dong-Hyun Lee
- Department of Obstetrics and Gynecology, Chonbuk National University Medical School, Jeonju, Korea
| | - Sang-Myeong Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea.
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Korea.
| |
Collapse
|
8
|
Nishi H, Niyonsaba F, Pelleg A, Schulman ES. Enhancement of Mast Cell Degranulation Mediated by Purinergic Receptors' Activation and PI3K Type δ. THE JOURNAL OF IMMUNOLOGY 2021; 207:1001-1008. [PMID: 34330752 DOI: 10.4049/jimmunol.2001002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 05/28/2021] [Indexed: 11/19/2022]
Abstract
Mast cells express multiple metabotropic purinergic P2Y receptor (P2YR) subtypes. Few studies have evaluated their role in human mast cell (HMC) allergic response as quantified by degranulation induced by cross-linking the high-affinity IgE receptor (FcεRI). We have previously shown that extracellular nucleotides modify the FcεRI activation-dependent degranulation in HMCs derived from human lungs, but the mechanism of this action has not been fully delineated. This study was undertaken to determine the mechanism of activation of P2YRs on the degranulation of HMCs and elucidate the specific postreceptor pathways involved. Sensitized LAD2 cells, a human-derived mast cell line, were subjected to a weak allergic stimulation (WAS) using a low concentration of Ag in the absence and presence of P2YR agonists. Only the metabotropic purinergic P2Y11 receptor (P2Y11R) agonist, adenosine 5'-(3-thio)triphosphate (ATPγS), enhanced WAS-induced degranulation resulting in a net 7-fold increase in release (n = 4; p < 0.01). None of the P2YR agonists tested, including high concentrations of ATPγS (1000 μM), enhanced WAS-induced intracellular Ca2+ mobilization, an essential component of activated FcεRI-induced degranulation. Both a PI3K inhibitor and the relevant gene knockout decreased the ATPγS-induced enhancement. The effect of ATPγS was associated with enhanced phosphorylation of PI3K type δ and protein kinase B, but not the phosphoinositide-dependent kinase-1. The effects of ATPγS were dose dependently inhibited by NF157, a P2Y11R antagonist. To our knowledge, these data indicate for the first time that P2YR is linked to enhancement of allergic degranulation in HMC via the PI3K/protein kinase B pathway.
Collapse
Affiliation(s)
- Haruhisa Nishi
- Department of Pharmacology, Jikei University School of Medicine, Tokyo, Japan;
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Faculty of International Liberal Arts, Juntendo University, Tokyo, Japan
| | - Amir Pelleg
- Danmir Therapeutics, LLC, Haverford, PA; and
| | | |
Collapse
|
9
|
Ménasché G, Longé C, Bratti M, Blank U. Cytoskeletal Transport, Reorganization, and Fusion Regulation in Mast Cell-Stimulus Secretion Coupling. Front Cell Dev Biol 2021; 9:652077. [PMID: 33796537 PMCID: PMC8007931 DOI: 10.3389/fcell.2021.652077] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/03/2021] [Indexed: 01/16/2023] Open
Abstract
Mast cells are well known for their role in allergies and many chronic inflammatory diseases. They release upon stimulation, e.g., via the IgE receptor, numerous bioactive compounds from cytoplasmic secretory granules. The regulation of granule secretion and its interaction with the cytoskeleton and transport mechanisms has only recently begun to be understood. These studies have provided new insight into the interaction between the secretory machinery and cytoskeletal elements in the regulation of the degranulation process. They suggest a tight coupling of these two systems, implying a series of specific signaling effectors and adaptor molecules. Here we review recent knowledge describing the signaling events regulating cytoskeletal reorganization and secretory granule transport machinery in conjunction with the membrane fusion machinery that occur during mast cell degranulation. The new insight into MC biology offers novel strategies to treat human allergic and inflammatory diseases targeting the late steps that affect harmful release from granular stores leaving regulatory cytokine secretion intact.
Collapse
Affiliation(s)
- Gaël Ménasché
- Laboratory of Molecular Basis of Altered Immune Homeostasis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Cyril Longé
- Laboratory of Molecular Basis of Altered Immune Homeostasis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Manuela Bratti
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, CNRS ERL8252, Faculté de Médecine site Bichat, Université de Paris, Paris, France.,Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Ulrich Blank
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, CNRS ERL8252, Faculté de Médecine site Bichat, Université de Paris, Paris, France.,Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| |
Collapse
|
10
|
Impact of Parenteral Lipid Emulsion Components on Cholestatic Liver Disease in Neonates. Nutrients 2021; 13:nu13020508. [PMID: 33557154 PMCID: PMC7913904 DOI: 10.3390/nu13020508] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/30/2022] Open
Abstract
Total parenteral nutrition (TPN) is a life-saving intervention for infants that are unable to feed by mouth. Infants that remain on TPN for extended periods of time are at risk for the development of liver injury in the form of parenteral nutrition associated cholestasis (PNAC). Current research suggests the lipid component of TPN is a factor in the development of PNAC. Most notably, the fatty acid composition, vitamin E concentration, and presence of phytosterols are believed key mediators of lipid emulsion driven PNAC development. New emulsions comprised of fish oil and medium chain triglycerides show promise for reducing the incidence of PNAC in infants. In this review we will cover the current clinical studies on the benefit of fish oil and medium chain triglyceride containing lipid emulsions on the development of PNAC, the current constituents of lipid emulsions that may modulate the prevalence of PNAC, and potential new supplements to TPN to further reduce the incidence of PNAC.
Collapse
|
11
|
Affiliation(s)
| | - Helen R. Mott
- Department of Biochemistry University of Cambridge Cambridge UK
| | - Darerca Owen
- Department of Biochemistry University of Cambridge Cambridge UK
| |
Collapse
|
12
|
Tsuge K, Inazumi T, Shimamoto A, Sugimoto Y. Molecular mechanisms underlying prostaglandin E2-exacerbated inflammation and immune diseases. Int Immunol 2020; 31:597-606. [PMID: 30926983 DOI: 10.1093/intimm/dxz021] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/18/2019] [Indexed: 01/12/2023] Open
Abstract
Prostaglandins (PGs) are the major lipid mediators in animals and which are biosynthesized from arachidonic acid by the cyclooxygenases (COX-1 or COX-2) as the rate-limiting enzymes. Prostaglandin E2 (PGE2), which is the most abundantly detected PG in various tissues, exerts versatile physiological and pathological actions via four receptor subtypes (EP1-4). Non-steroidal anti-inflammatory drugs, such as aspirin and indomethacin, exert potent anti-inflammatory actions by the inhibition of COX activity and the resulting suppression of PG production. Therefore, PGE2 has been shown to exacerbate several inflammatory responses and immune diseases. Recently, studies using mice deficient in each PG receptor subtype have clarified the detailed mechanisms underlying PGE2-associated inflammation and autoimmune diseases involving each EP receptor. Here, we review the recent advances in our understanding of the roles of PGE2 receptors in the progression of acute and chronic inflammation and autoimmune diseases. PGE2 induces acute inflammation through mast cell activation via the EP3 receptor. PGE2 also induces chronic inflammation and various autoimmune diseases through T helper 1 (Th1)-cell differentiation, Th17-cell proliferation and IL-22 production from Th22 cells via the EP2 and EP4 receptors. The possibility of EP receptor-targeted drug development for the treatment of immune diseases is also discussed.
Collapse
Affiliation(s)
- Kyoshiro Tsuge
- Department of Regenerative Medicine Research, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda, Yamaguchi, Japan
| | - Tomoaki Inazumi
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology, Chiyoda-ku, Tokyo, Japan
| | - Akira Shimamoto
- Department of Regenerative Medicine Research, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda, Yamaguchi, Japan
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
13
|
Bryson TD, Pandrangi TS, Khan SZ, Xu J, Pavlov TS, Ortiz PA, Peterson E, Harding P. The deleterious role of the prostaglandin E 2 EP 3 receptor in angiotensin II hypertension. Am J Physiol Heart Circ Physiol 2020; 318:H867-H882. [PMID: 32142358 DOI: 10.1152/ajpheart.00538.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiotensin II (ANG II) plays a key role in regulating blood pressure and inflammation. Prostaglandin E2 (PGE2) signals through four different G protein-coupled receptors, eliciting a variety of effects. We reported that activation of the EP3 receptor reduces cardiac contractility. More recently, we have shown that overexpression of the EP4 receptor is protective in a mouse myocardial infarction model. We hypothesize in this study that the relative abundance of EP3 and EP4 receptors is a major determinant of end-organ damage in the diseased heart. Thus EP3 is detrimental to cardiac function and promotes inflammation, whereas antagonism of the EP3 receptor is protective in an ANG II hypertension (HTN) model. To test our hypothesis, male 10- to 12-wk-old C57BL/6 mice were anesthetized with isoflurane and osmotic minipumps containing ANG II were implanted subcutaneously for 2 wk. We found that antagonism of the EP3 receptor using L798,106 significantly attenuated the increase in blood pressure with ANG II infusion. Moreover, antagonism of the EP3 receptor prevented a decline in cardiac function after ANG II treatment. We also found that 10- to 12-wk-old EP3-transgenic mice, which overexpress EP3 in the cardiomyocytes, have worsened cardiac function. In conclusion, activation or overexpression of EP3 exacerbates end-organ damage in ANG II HTN. In contrast, antagonism of the EP3 receptor is beneficial and reduces cardiac dysfunction, inflammation, and HTN.NEW & NOTEWORTHY This study is the first to show that systemic treatment with an EP3 receptor antagonist (L798,106) attenuates the angiotensin II-induced increase in blood pressure in mice. The results from this project could complement existing hypertension therapies by combining blockade of the EP3 receptor with antihypertensive drugs.
Collapse
Affiliation(s)
- Timothy D Bryson
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Teja S Pandrangi
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Safa Z Khan
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Jiang Xu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Tengis S Pavlov
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Edward Peterson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan
| | - Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
14
|
Trigonelline: An alkaloid with anti-degranulation properties. Mol Immunol 2020; 118:201-209. [DOI: 10.1016/j.molimm.2019.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 01/23/2023]
|
15
|
Park YH, Kim DK, Kim HS, Lee D, Lee MB, Min KY, Jo MG, Lee JE, Kim YM, Choi WS. WZ3146 inhibits mast cell Lyn and Fyn to reduce IgE-mediated allergic responses in vitro and in vivo. Toxicol Appl Pharmacol 2019; 383:114763. [PMID: 31526816 DOI: 10.1016/j.taap.2019.114763] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/06/2019] [Accepted: 09/13/2019] [Indexed: 11/19/2022]
Abstract
Mast cells (MCs) play an important role as effector cells that cause allergic responses in allergic diseases. For these reasons, MC is considered an attractive therapeutic target for allergic disease treatment. In this study, we investigated the inhibitory effect of WZ3146, N-[3-[5-chloro-2-[4-(4-methylpiperazin-1-yl)anilino]pyrimidin-4-yl]oxyphenyl]prop-2-enamide, and the mechanisms of its actions on the MC activation and IgE-mediated allergic response by using three types of MCs such as rat basophilic leukemia (RBL)-2H3 cells, mouse bone marrow mast cells (BMMCs), and human Laboratory of Allergic Diseases 2 (LAD2) cells. WZ3146 inhibited antigen-stimulated degranulation in a dose-dependent manner (IC50, ~ 0.35 μM for RBL-2H3 cells; ~ 0.39 μM for BMMCs; ~ 0.41 for LAD2 cells). WZ3146 also suppressed the production of histamine, tumor necrosis factor (TNF)-α and interleukin (IL)-6, which mediate various allergic responses, in a dose-dependent manner. As the mechanism of WZ3146 to inhibit MCs, it inhibited the activation of spleen tyrosine kinase (Syk) and the downstream signaling proteins of Syk such as linker for activation of T cell (LAT) and phospholipase (PL) Cγ1 in the signaling pathway of FcεRI. In addition, WZ3146 inhibited the activation of Akt, extracellular signal-regulated kinase (ERK)1/2, p38, and c-Jun N-terminal kinase (JNK). However, WZ3146 did not inhibit degranulation of MCs by thapsigargin or ionomycin, which increase calcium concentration in cytosol. Notably, WZ3146 inhibited the activity of Lyn and Fyn, but not Syk. In an following animal experiment, WZ3146 inhibited IgE-dependent passive cutaneous anaphylaxis (PCA) in a dose-dependent manner (ED50, ~ 20 mg/kg). Taken together, in this study we show that the pyrimidine derivative, WZ3146, inhibits the IgE-mediated allergic response by inhibiting Lyn and Fyn Src-family kinases, which are initially activated by antigen stimulation in MCs. Therefore, we propose that WZ3146 could be used as a new therapeutic agent for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Young Hwan Park
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Do Kyun Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Hyuk Soon Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Dajeong Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Bum Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Geun Jo
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Ji Eon Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea.
| |
Collapse
|
16
|
Uchida R, Egawa T, Fujita Y, Furuta K, Taguchi H, Tanaka S, Nishida K. Identification of the minimal region of peptide derived from ADP-ribosylation factor1 (ARF1) that inhibits IgE-mediated mast cell activation. Mol Immunol 2019; 105:32-37. [DOI: 10.1016/j.molimm.2018.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 10/23/2018] [Accepted: 11/08/2018] [Indexed: 10/27/2022]
|
17
|
Park YH, Kim DK, Kim HW, Kim HS, Lee D, Lee MB, Min KY, Koo J, Kim SJ, Kang C, Kim YM, Kim HS, Choi WS. Repositioning of anti-cancer drug candidate, AZD7762, to an anti-allergic drug suppressing IgE-mediated mast cells and allergic responses via the inhibition of Lyn and Fyn. Biochem Pharmacol 2018; 154:270-277. [PMID: 29777684 DOI: 10.1016/j.bcp.2018.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/15/2018] [Indexed: 01/26/2023]
Abstract
Mast cells are critical effector cells in IgE-mediated allergic responses. The aim of this study was to investigate the anti-allergic effects of 3-[(aminocarbonyl)amino]-5-(3-fluorophenyl)-N-(3S)-3-piperidinyl-2-thiophenecarboxamide (AZD7762) in vitro and in vivo. AZD7762 inhibited the antigen-stimulated degranulation from RBL-2H3 (IC50, ∼27.9 nM) and BMMCs (IC50, ∼99.3 nM) in a dose-dependent manner. AZD7762 also inhibited the production of TNF-α and IL-4. As the mechanism of its action, AZD7762 inhibited the activation of Syk and its downstream signaling proteins, such as Linker of activated T cells (LAT), phospholipase (PL) Cγ1, Akt, and mitogen-activated protein (MAP) kinases (Erk1/2, p38, and JNK) in mast cells. In in vitro protein kinase assay, AZD7762 inhibited the activity of Lyn and Fyn kinases, which are important for the activation of Syk in mast cells. Furthermore, AZD7762 also suppressed the degranulation of LAD2 human mast cells (IC50, ∼49.9 nM) and activation of Syk in a dose-dependent manner. As observed in experiments with mast cells in vitro, AZD7762 inhibited antigen-mediated passive cutaneous anaphylaxis in mice (ED50, ∼35.8 mg/kg). Altogether, these results suggest that AZD7762 could be used as a new therapeutic agent for mast cell-mediated allergic diseases.
Collapse
Affiliation(s)
- Young Hwan Park
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Do-Kyun Kim
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Hyun Woo Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Hyuk Soon Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Dajeong Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Bum Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Jimo Koo
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Su Jeong Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Changhee Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Hyung Sik Kim
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea.
| |
Collapse
|
18
|
Munoz I, Danelli L, Claver J, Goudin N, Kurowska M, Madera-Salcedo IK, Huang JD, Fischer A, González-Espinosa C, de Saint Basile G, Blank U, Ménasché G. Kinesin-1 controls mast cell degranulation and anaphylaxis through PI3K-dependent recruitment to the granular Slp3/Rab27b complex. J Cell Biol 2017; 215:203-216. [PMID: 27810912 PMCID: PMC5084650 DOI: 10.1083/jcb.201605073] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/12/2016] [Indexed: 12/22/2022] Open
Abstract
Activation of mast cells through IgE and antigen triggers the release of secretory granules that contain factors responsible for anaphylactic responses. Munoz et al. show that kinesin-1 regulates mast cell degranulation through PI3K-dependent formation of a kinesin-1/Slp3/Rab27b complex. Cross-linking of mast cell (MC) IgE receptors (FcεRI) triggers degranulation of secretory granules (SGs) and the release of many allergic and inflammatory mediators. Although degranulation depends crucially on microtubule dynamics, the molecular machinery that couples SGs to microtubule-dependent transport is poorly understood. In this study, we demonstrate that mice lacking Kif5b (the heavy chain of kinesin-1) in hematopoietic cells are less sensitive to IgE-mediated, passive, systemic anaphylaxis. After IgE-induced stimulation, bone marrow–derived MCs from Kif5b knockout mice exhibited a marked reduction in SG translocation toward the secretion site. In contrast, a lack of Kif5b did not affect cytokine secretion, early FcεRI-initiated signaling pathways, or microtubule reorganization upon FcεRI stimulation. We identified Slp3 as the critical effector linking kinesin-1 to Rab27b-associated SGs. Kinesin-1 recruitment to the Slp3/Rab27b effector complex was independent of microtubule reorganization but occurred only upon stimulation requiring phosphatidylinositol 3-kinase (PI3K) activity. Our findings demonstrate that PI3K-dependent formation of a kinesin-1/Slp3/Rab27b complex is critical for the microtubule-dependent movement of SGs required for MC degranulation.
Collapse
Affiliation(s)
- Isabelle Munoz
- Laboratory of Normal and Pathological Homeostasis of the Immune System, Institut National de la Santé et de la Recherche Médicale, UMR1163, F-75015 Paris, France.,Imagine Institute, Paris Descartes University-Sorbonne Paris Cité, F-75015 Paris, France
| | - Luca Danelli
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1149; Centre de Recherche sur l'Inflammation, Centre National de la Recherche Scientifique, Equipe de Recherche Labelisé 8252; Inflamex Laboratory of Excellence, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France
| | - Julien Claver
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1149; Centre de Recherche sur l'Inflammation, Centre National de la Recherche Scientifique, Equipe de Recherche Labelisé 8252; Inflamex Laboratory of Excellence, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France
| | - Nicolas Goudin
- Cell Imaging Facility, Imagine Institute, Paris Descartes University-Sorbonne Paris Cité, F-75015 Paris, France
| | - Mathieu Kurowska
- Laboratory of Normal and Pathological Homeostasis of the Immune System, Institut National de la Santé et de la Recherche Médicale, UMR1163, F-75015 Paris, France.,Imagine Institute, Paris Descartes University-Sorbonne Paris Cité, F-75015 Paris, France
| | - Iris Karina Madera-Salcedo
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1149; Centre de Recherche sur l'Inflammation, Centre National de la Recherche Scientifique, Equipe de Recherche Labelisé 8252; Inflamex Laboratory of Excellence, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France
| | - Jian-Dong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Alain Fischer
- Laboratory of Normal and Pathological Homeostasis of the Immune System, Institut National de la Santé et de la Recherche Médicale, UMR1163, F-75015 Paris, France.,Imagine Institute, Paris Descartes University-Sorbonne Paris Cité, F-75015 Paris, France.,Immunology and Pediatric Hematology Department, Necker Children's Hospital, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France.,Collège de France, F-75005 Paris, France
| | | | - Geneviéve de Saint Basile
- Laboratory of Normal and Pathological Homeostasis of the Immune System, Institut National de la Santé et de la Recherche Médicale, UMR1163, F-75015 Paris, France.,Imagine Institute, Paris Descartes University-Sorbonne Paris Cité, F-75015 Paris, France
| | - Ulrich Blank
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1149; Centre de Recherche sur l'Inflammation, Centre National de la Recherche Scientifique, Equipe de Recherche Labelisé 8252; Inflamex Laboratory of Excellence, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France
| | - Gaël Ménasché
- Laboratory of Normal and Pathological Homeostasis of the Immune System, Institut National de la Santé et de la Recherche Médicale, UMR1163, F-75015 Paris, France .,Imagine Institute, Paris Descartes University-Sorbonne Paris Cité, F-75015 Paris, France
| |
Collapse
|
19
|
Extracellular Signal-Regulated Kinases 1 and 2 Phosphorylate Gab2 To Promote a Negative-Feedback Loop That Attenuates Phosphoinositide 3-Kinase/Akt Signaling. Mol Cell Biol 2017; 37:MCB.00357-16. [PMID: 28096188 DOI: 10.1128/mcb.00357-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 12/28/2016] [Indexed: 11/20/2022] Open
Abstract
The scaffolding adapter protein Gab2 (Grb2-associated binder) promotes cell proliferation, survival, and motility by engaging several signaling pathways downstream of growth factor and cytokine receptors. In particular, Gab2 plays essential roles in mast cells, as it is required for phosphoinositide 3-kinase (PI3K) activation in response to Kit and the high-affinity IgE receptor. While the positive role of Gab2 in PI3K signaling is well documented, very little is known about the mechanisms that attenuate its function. Here we show that Gab2 becomes phosphorylated on multiple proline-directed sites upon stimulation of the Ras/extracellular signal-regulated kinase (ERK) signaling pathway. We demonstrate that ERK1 and ERK2 interact with Gab2 via a novel docking motif, which is required for subsequent Gab2 phosphorylation in response to ERK1/2 activation. We identified four ERK1/2-dependent phosphorylation sites in Gab2 that prevent the recruitment of the p85 regulatory subunit of PI3K. Using bone marrow-derived mast cells to study Gab2-dependent signaling, we found that the inhibition of ERK1/2 activity promotes Akt signaling in response to Kit and the high-affinity IgE receptor. Together, our results indicate that ERK1/2 participates in a negative-feedback loop that attenuates PI3K/Akt signaling in response to various agonists.
Collapse
|
20
|
Khadilkar RJ, Ray A, Chetan DR, Sinha AR, Magadi SS, Kulkarni V, Inamdar MS. Differential modulation of the cellular and humoral immune responses in Drosophila is mediated by the endosomal ARF1-Asrij axis. Sci Rep 2017; 7:118. [PMID: 28273919 PMCID: PMC5427928 DOI: 10.1038/s41598-017-00118-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022] Open
Abstract
How multicellular organisms maintain immune homeostasis across various organs and cell types is an outstanding question in immune biology and cell signaling. In Drosophila, blood cells (hemocytes) respond to local and systemic cues to mount an immune response. While endosomal regulation of Drosophila hematopoiesis is reported, the role of endosomal proteins in cellular and humoral immunity is not well-studied. Here we demonstrate a functional role for endosomal proteins in immune homeostasis. We show that the ubiquitous trafficking protein ADP Ribosylation Factor 1 (ARF1) and the hemocyte-specific endosomal regulator Asrij differentially regulate humoral immunity. Asrij and ARF1 play an important role in regulating the cellular immune response by controlling the crystal cell melanization and phenoloxidase activity. ARF1 and Asrij mutants show reduced survival and lifespan upon infection, indicating perturbed immune homeostasis. The ARF1-Asrij axis suppresses the Toll pathway anti-microbial peptides (AMPs) by regulating ubiquitination of the inhibitor Cactus. The Imd pathway is inversely regulated- while ARF1 suppresses AMPs, Asrij is essential for AMP production. Several immune mutants have reduced Asrij expression, suggesting that Asrij co-ordinates with these pathways to regulate the immune response. Our study highlights the role of endosomal proteins in modulating the immune response by maintaining the balance of AMP production. Similar mechanisms can now be tested in mammalian hematopoiesis and immunity.
Collapse
Affiliation(s)
- Rohan J Khadilkar
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Arindam Ray
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - D R Chetan
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | | | - Srivathsa S Magadi
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Vani Kulkarni
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Maneesha S Inamdar
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.
| |
Collapse
|
21
|
Gu G, Chen Y, Duan C, Zhou L, Chen C, Chen J, Cheng J, Shi N, Jin Y, Xi Q, Zhong J. Overexpression of ARF1 is associated with cell proliferation and migration through PI3K signal pathway in ovarian cancer. Oncol Rep 2017; 37:1511-1520. [DOI: 10.3892/or.2017.5388] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/15/2016] [Indexed: 11/06/2022] Open
|
22
|
Abdel-Rahman SM. Genetic Predictors of Susceptibility to Dermatophytoses. Mycopathologia 2016; 182:67-76. [PMID: 27502504 DOI: 10.1007/s11046-016-0046-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 07/27/2016] [Indexed: 12/27/2022]
Abstract
Countless observational studies conducted over the last century reveal that dermatophytes infect humans of every age, race, gender, and socioeconomic status with strikingly high rates. The curious disparity in dermatophyte infection patterns observed within and between populations has led countless investigators to explore whether genetics underlie a susceptibility to, or confer protection against, dermatophyte infections. This paper examines the data that offer a link between genetics and dermatophytoses and discusses the underlying mechanisms that support these observations.
Collapse
Affiliation(s)
- Susan M Abdel-Rahman
- UMKC School of Medicine, Kansas City, MO, USA. .,Section of Therapeutic Innovation, Children's Mercy Hospital, Kansas City, MO, USA. .,Division of Pediatric Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy Hospitals and Clinics, 2401 Gillham Rd., Kansas City, MO, 64108, USA.
| |
Collapse
|
23
|
Yashin AI, Arbeev KG, Wu D, Arbeeva L, Kulminski A, Kulminskaya I, Akushevich I, Ukraintseva SV. How Genes Modulate Patterns of Aging-Related Changes on the Way to 100: Biodemographic Models and Methods in Genetic Analyses of Longitudinal Data. NORTH AMERICAN ACTUARIAL JOURNAL : NAAJ 2016; 20:201-232. [PMID: 27773987 PMCID: PMC5070546 DOI: 10.1080/10920277.2016.1178588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVE To clarify mechanisms of genetic regulation of human aging and longevity traits, a number of genome-wide association studies (GWAS) of these traits have been performed. However, the results of these analyses did not meet expectations of the researchers. Most detected genetic associations have not reached a genome-wide level of statistical significance, and suffered from the lack of replication in the studies of independent populations. The reasons for slow progress in this research area include low efficiency of statistical methods used in data analyses, genetic heterogeneity of aging and longevity related traits, possibility of pleiotropic (e.g., age dependent) effects of genetic variants on such traits, underestimation of the effects of (i) mortality selection in genetically heterogeneous cohorts, (ii) external factors and differences in genetic backgrounds of individuals in the populations under study, the weakness of conceptual biological framework that does not fully account for above mentioned factors. One more limitation of conducted studies is that they did not fully realize the potential of longitudinal data that allow for evaluating how genetic influences on life span are mediated by physiological variables and other biomarkers during the life course. The objective of this paper is to address these issues. DATA AND METHODS We performed GWAS of human life span using different subsets of data from the original Framingham Heart Study cohort corresponding to different quality control (QC) procedures and used one subset of selected genetic variants for further analyses. We used simulation study to show that approach to combining data improves the quality of GWAS. We used FHS longitudinal data to compare average age trajectories of physiological variables in carriers and non-carriers of selected genetic variants. We used stochastic process model of human mortality and aging to investigate genetic influence on hidden biomarkers of aging and on dynamic interaction between aging and longevity. We investigated properties of genes related to selected variants and their roles in signaling and metabolic pathways. RESULTS We showed that the use of different QC procedures results in different sets of genetic variants associated with life span. We selected 24 genetic variants negatively associated with life span. We showed that the joint analyses of genetic data at the time of bio-specimen collection and follow up data substantially improved significance of associations of selected 24 SNPs with life span. We also showed that aging related changes in physiological variables and in hidden biomarkers of aging differ for the groups of carriers and non-carriers of selected variants. CONCLUSIONS . The results of these analyses demonstrated benefits of using biodemographic models and methods in genetic association studies of these traits. Our findings showed that the absence of a large number of genetic variants with deleterious effects may make substantial contribution to exceptional longevity. These effects are dynamically mediated by a number of physiological variables and hidden biomarkers of aging. The results of these research demonstrated benefits of using integrative statistical models of mortality risks in genetic studies of human aging and longevity.
Collapse
Affiliation(s)
- Anatoliy I. Yashin
- Professor, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A102E, Durham, NC 27705, USA. Tel.: (+1) 919-668-2713; Fax: (+1) 919-684-3861
| | - Konstantin G. Arbeev
- Sr. Research Scientist, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A102F, Durham, NC 27705, USA. Tel.: (+1) 919-668-2707; Fax: (+1) 919-684-3861
| | - Deqing Wu
- Sr. Research Scientist, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A104, Durham, NC 27705, USA. Tel.: (+1) 919-684-6126; Fax: (+1) 919-684-3861
| | - Liubov Arbeeva
- Statistician, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A102G, Durham, NC 27705, USA. Tel.: (+1) 919-613-0715; Fax: (+1) 919-684-3861
| | - Alexander Kulminski
- Sr. Research Scientist, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A106, Durham, NC 27705, USA. Tel.: (+1) 919-684-4962; Fax: (+1) 919-684-3861
| | - Irina Kulminskaya
- Research Scientist, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A102D, Durham, NC 27705, USA. Tel.: (+1) 919-681-8232; Fax: (+1) 919-684-3861
| | - Igor Akushevich
- Sr. Research Scientist, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A107, Durham, NC 27705, USA. Tel.: (+1) 919-668-2715; Fax: (+1) 919-684-3861
| | - Svetlana V. Ukraintseva
- Sr. Research Scientist, Center for Population Health and Aging, Duke University, 2024 W. Main Street, Room A105, Durham, NC 27705, USA. Tel.: (+1) 919-668-2712; Fax: (+1) 919-684-3861
| |
Collapse
|
24
|
Kawahara K, Hohjoh H, Inazumi T, Tsuchiya S, Sugimoto Y. Prostaglandin E2-induced inflammation: Relevance of prostaglandin E receptors. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:414-21. [PMID: 25038274 DOI: 10.1016/j.bbalip.2014.07.008] [Citation(s) in RCA: 307] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/04/2014] [Accepted: 07/10/2014] [Indexed: 12/15/2022]
Abstract
Prostaglandin E2 (PGE2) is one of the most typical lipid mediators produced from arachidonic acid (AA) by cyclooxygenase (COX) as the rate-limiting enzyme, and acts on four kinds of receptor subtypes (EP1-EP4) to elicit its diverse actions including pyrexia, pain sensation, and inflammation. Recently, the molecular mechanisms underlying the PGE2 actions mediated by each EP subtype have been elucidated by studies using mice deficient in each EP subtype as well as several compounds highly selective to each EP subtype, and their findings now enable us to discuss how PGE2 initiates and exacerbates inflammation at the molecular level. Here, we review the recent advances in PGE2 receptor research by focusing on the activation of mast cells via the EP3 receptor and the control of helper T cells via the EP2/4 receptor, which are the molecular mechanisms involved in PGE2-induced inflammation that had been unknown for many years. We also discuss the roles of PGE2 in acute inflammation and inflammatory disorders, and the usefulness of anti-inflammatory therapies that target EP receptors. This article is part of a Special Issue entitled "Oxygenated metabolism of PUFA: analysis and biological relevance".
Collapse
Affiliation(s)
- Kohichi Kawahara
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Hirofumi Hohjoh
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Tomoaki Inazumi
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Soken Tsuchiya
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan.
| |
Collapse
|
25
|
Ogawa K, Tanaka Y, Uruno T, Duan X, Harada Y, Sanematsu F, Yamamura K, Terasawa M, Nishikimi A, Côté JF, Fukui Y. DOCK5 functions as a key signaling adaptor that links FcεRI signals to microtubule dynamics during mast cell degranulation. ACTA ACUST UNITED AC 2014; 211:1407-19. [PMID: 24913231 PMCID: PMC4076576 DOI: 10.1084/jem.20131926] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Mast cells play a key role in the induction of anaphylaxis, a life-threatening IgE-dependent allergic reaction, by secreting chemical mediators that are stored in secretory granules. Degranulation of mast cells is triggered by aggregation of the high-affinity IgE receptor, FcεRI, and involves dynamic rearrangement of microtubules. Although much is known about proximal signals downstream of FcεRI, the distal signaling events controlling microtubule dynamics remain elusive. Here we report that DOCK5, an atypical guanine nucleotide exchange factor (GEF) for Rac, is essential for mast cell degranulation. As such, we found that DOCK5-deficient mice exhibit resistance to systemic and cutaneous anaphylaxis. The Rac GEF activity of DOCK5 is surprisingly not required for mast cell degranulation. Instead, DOCK5 associated with Nck2 and Akt to regulate microtubule dynamics through phosphorylation and inactivation of GSK3β. When DOCK5-Nck2-Akt interactions were disrupted, microtubule formation and degranulation response were severely impaired. Our results thus identify DOCK5 as a key signaling adaptor that orchestrates remodeling of the microtubule network essential for mast cell degranulation.
Collapse
Affiliation(s)
- Kana Ogawa
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Yoshihiko Tanaka
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Takehito Uruno
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Xuefeng Duan
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Yosuke Harada
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Fumiyuki Sanematsu
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Kazuhiko Yamamura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Masao Terasawa
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Akihiko Nishikimi
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Quebec H2W 1R7, Canada
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| |
Collapse
|
26
|
Morimoto K, Shirata N, Taketomi Y, Tsuchiya S, Segi-Nishida E, Inazumi T, Kabashima K, Tanaka S, Murakami M, Narumiya S, Sugimoto Y. Prostaglandin E2–EP3 Signaling Induces Inflammatory Swelling by Mast Cell Activation. THE JOURNAL OF IMMUNOLOGY 2013; 192:1130-7. [DOI: 10.4049/jimmunol.1300290] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
27
|
Chan TK, Ng DSW, Cheng C, Guan SP, Koh HM, Wong WSF. Anti-allergic actions of rottlerin from Mallotus philippinensis in experimental mast cell-mediated anaphylactic models. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:853-860. [PMID: 23632085 DOI: 10.1016/j.phymed.2013.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/04/2013] [Accepted: 03/09/2013] [Indexed: 05/28/2023]
Abstract
Allergy is an acquired hypersensitivity reaction of the immune system mediated by cross-linking of the allergen-specific IgE-bound high-affinity IgE receptors, leading to immediate mast cell degranulation. Rottlerin is an active molecule isolated from Mallotus philippinensis, a medicinal plant used in Ayurvedic Medicine System for anti-allergic and anti-helminthic treatments. The present study investigated potential anti-allergic effects of rottlerin in animal models of IgE-dependent anaphylaxis and the anti-allergic mechanisms of action of rottlerin in mast cells. Anti-allergic actions of rottlerin were evaluated in passive cutaneous anaphylaxis and passive systemic anaphylaxis mouse models, and in anaphylactic contraction of bronchial rings isolated from sensitized guinea pigs. Direct mast cell-stabilizing effect of rottlerin was examined in RBL-2H3 mast cell line. Anti-allergic signaling mechanisms of action of rottlerin in mast cells were also examined. Rottlerin prevented IgE-mediated cutaneous vascular extravasation, hypothermia, elevation in plasma histamine level and tracheal tissue mast cell degranulation in mice in a dose-dependent manner. In addition, rottlerin suppressed ovalbumin-induced guinea pig bronchial smooth muscle contraction. Furthermore, rottlerin concentration-dependently blocked IgE-mediated immediate release of β-hexosaminidase from RBL-2H3 mast cells. Rottlerin was found to inhibit IgE-induced PLCγ1 and Akt phosphorylation, production of IP3 and rise in cytosolic Ca²⁺ level in mast cells. We report here for the first time that rottlerin possesses anti-allergic activity by blocking IgE-induced mast cell degranulation, providing a foundation for developing rottlerin for the treatment of allergic asthma and other mast cell-mediated allergic disorders.
Collapse
Affiliation(s)
- Tze Khee Chan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | | | | | | | | | | |
Collapse
|
28
|
Cruse G, Beaven MA, Ashmole I, Bradding P, Gilfillan AM, Metcalfe DD. A truncated splice-variant of the FcεRIβ receptor subunit is critical for microtubule formation and degranulation in mast cells. Immunity 2013; 38:906-17. [PMID: 23643722 DOI: 10.1016/j.immuni.2013.04.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 01/15/2013] [Indexed: 01/25/2023]
Abstract
Human linkage analyses have implicated the MS4A2-containing gene locus (encoding FcεRIβ) as a candidate for allergy susceptibility. We have identified a truncation of FcεRIβ (t-FcεRIβ) in humans that contains a putative calmodulin-binding domain and thus, we sought to identify the role of this variant in mast cell function. We determined that t-FcεRIβ is critical for microtubule formation and degranulation and that it may perform this function by trafficking adaptor molecules and kinases to the pericentrosomal and Golgi region in response to Ca2+ signals. Mutagenesis studies suggest that calmodulin binding to t-FcεRIβ in the presence of Ca2+ could be critical for t-FcεRIβ function. In addition, gene targeting of t-FcεRIβ attenuated microtubule formation, degranulation, and IL-8 production downstream of Ca2+ signals. Therefore, t-FcεRIβ mediates Ca2+ -dependent microtubule formation, which promotes degranulation and cytokine release. Because t-FcεRIβ has this critical function, it represents a therapeutic target for the downregulation of allergic inflammation.
Collapse
Affiliation(s)
- Glenn Cruse
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
A range of mediators are generated during anaphylaxis, with redundancy of effects, multiple overlapping pathways, and involvement of several cell types. Key steps in the reaction occur at the site of initial contact, and mediators may not be detectable systemically. Furthermore, the potencies of various mediators vary enormously, and clinical effects may occur below our level of detection. We also do not know what converts (amplifies) a local reaction into systemic anaphylaxis. Murine models have identified several novel mediators that may propagate and/or regulate this process and also indicate that circulating neutrophils may play an important role in reaction amplification. Differential expression of various genes within specific intracellular signalling pathways of mediator release may further explain the varying severities of anaphylactic reactions. As our knowledge of the mechanisms of activation, key mediators, and the regulation of mediator release improves, new treatments for prevention and acute management may emerge.
Collapse
Affiliation(s)
- Shelley F Stone
- Centre for Clinical Research in Emergency Medicine, Western Australian Institute for Medical Research, University of Western Australia, Perth, Western Australia, Australia.
| | | |
Collapse
|
30
|
Gab docking proteins in cardiovascular disease, cancer, and inflammation. Int J Inflam 2013; 2013:141068. [PMID: 23431498 PMCID: PMC3566608 DOI: 10.1155/2013/141068] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 12/11/2012] [Indexed: 12/23/2022] Open
Abstract
The docking proteins of the Grb2-associated binder (Gab) family have emerged as crucial signaling compartments in metazoans. In mammals, the Gab proteins, consisting of Gab1, Gab2, and Gab3, are involved in the amplification and integration of signal transduction evoked by a variety of extracellular stimuli, including growth factors, cytokines, antigens, and other molecules. Gab proteins lack the enzymatic activity themselves; however, when phosphorylated on tyrosine residues, they provide binding sites for multiple Src homology-2 (SH2) domain-containing proteins, such as SH2-containing protein tyrosine phosphatase 2 (SHP2), phosphatidylinositol 3-kinase regulatory subunit p85, phospholipase Cγ, Crk, and GC-GAP. Through these interactions, the Gab proteins transduce signals from activated receptors into pathways with distinct biological functions, thereby contributing to signal diversification. They are known to play crucial roles in numerous physiological processes through their associations with SHP2 and p85. In addition, abnormal Gab protein signaling has been linked to human diseases including cancer, cardiovascular disease, and inflammatory disorders. In this paper, we provide an overview of the structure, effector functions, and regulation of the Gab docking proteins, with a special focus on their associations with cardiovascular disease, cancer, and inflammation.
Collapse
|
31
|
Nishida K. [New knowledge from past decade: role of zinc in immune system]. Nihon Eiseigaku Zasshi 2013; 68:145-152. [PMID: 24077486 DOI: 10.1265/jjh.68.145] [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: 06/02/2023]
Abstract
Zinc (Zn) is essential for normal cell structure and physiology. Its deficiency causes growth retardation, neuronal degeneration, and immunodeficiency. Zn homeostasis is tightly controlled through Zn transporters and metallothioneins, which regulate Zn concentration and Zn distribution in individual cells, and contributes to Zn-binding protein in cells. Although many molecules involved in these processes have Zn-binding motifs, the molecular mechanisms underlying the role of Zn in the immune system have not been clarified. Recently, we and other groups have demonstrated that Zn plays diverse and specific roles in vivo and in vitro, in studies on the genetic knockout of Zn transporter functions. In this review, we discuss the impact of Zn on mast cell-mediated allergy and T cell-mediated immune responses. We also describe Zn dysregulation as a leading health problem in allergy and immune responses.
Collapse
Affiliation(s)
- Keigo Nishida
- Laboratory for Homeostatic Network, RIKEN Center for Integrative Medial Sciences (IMS-RCAI)
| |
Collapse
|
32
|
Takayama G, Ohtani M, Minowa A, Matsuda S, Koyasu S. Class I PI3K-mediated Akt and ERK signals play a critical role in FcεRI-induced degranulation in mast cells. Int Immunol 2012; 25:215-20. [PMID: 23143475 DOI: 10.1093/intimm/dxs105] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Class IA and IB phosphoinositide 3-kinases (PI3Ks) have been shown to regulate mast cell functions such as proliferation, development, survival and degranulation, but the functional redundancy between these two PI3K signaling pathways in mast cells remains unclear. Here, we have generated mice deficient in both class IA regulatory subunit p85α and class IB catalytic subunit p110γ, and show that p85α(-/-)p110γ(-/-) mice exhibit a more severe defect in mast cell development than single-knockout mice. In addition, the in vivo passive cutaneous anaphylaxis reaction of p85α(-/-)p110γ(-/-) mice was nearly completely abrogated, whereas single-knockout mice exhibit just marginal reduction. Pharmacological inactivation of Akt in wild-type bone marrow-derived mast cells (BMMCs) led to partial reduction of degranulation, while over-expression of a constitutively active Akt partially restored the impaired degranulation in p85α(-/-)p110γ(-/-) BMMCs. We also found that the extracellular signal-regulated kinase (ERK) signaling pathway was activated in a PI3K-dependent manner upon FcεRI stimulation and that simultaneous inhibition of Akt and ERK resulted in nearly complete blockade of FcεRI-induced degranulation. Our data provide evidence that Akt and ERK pathways play redundant roles in FcεRI-induced degranulation.
Collapse
Affiliation(s)
- Gensuke Takayama
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
33
|
Kim W, Kim SY, Kim T, Kim M, Bae DJ, Choi HI, Kim IS, Jho E. ADP-ribosylation factors 1 and 6 regulate Wnt/β-catenin signaling via control of LRP6 phosphorylation. Oncogene 2012; 32:3390-6. [PMID: 22907437 DOI: 10.1038/onc.2012.373] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 06/26/2012] [Accepted: 07/13/2012] [Indexed: 12/25/2022]
Abstract
It has been shown that inhibition of GTPase-activating protein of ADP-ribosylation factor (Arf), ArfGAP, with a small molecule (QS11) results in synergistic activation of Wnt/β-catenin signaling. However, the role of Arf in Wnt/β-catenin signaling has not yet been elucidated. Here, we show that activation of Arf is essential for Wnt/β-catenin signaling. The level of the active form of Arf (Arf-GTP) transiently increased in the presence of Wnt, and this induction event was abrogated by blocking the interaction between Wnt and Frizzled (Fzd). In addition, knockdown of Fzds, Dvls or LRP6 blocked the Wnt-mediated activation of Arf. Consistently, depletion of Arf led to inhibition of Wnt-mediated membrane PtdIns (4,5)P2 (phosphatidylinositol 4, 5-bisphosphate) synthesis and LRP6 phosphorylation. Overall, our data suggest that transient activation of Arf modulates LRP6 phosphorylation for the transduction of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- W Kim
- Department of Life Science, The University of Seoul, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Fang X, Lang Y, Wang Y, Mo W, Wei H, Xie J, Yu M. Shp2 activates Fyn and Ras to regulate RBL-2H3 mast cell activation following FcεRI aggregation. PLoS One 2012; 7:e40566. [PMID: 22802969 PMCID: PMC3393662 DOI: 10.1371/journal.pone.0040566] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 06/09/2012] [Indexed: 11/18/2022] Open
Abstract
The protein-tyrosine phosphatase (PTP) Shp2 has been implicated in many immunoreceptor signaling pathways, but its role in immunoreceptor FcεRI signaling, which leads to the activation of mast cells and blood basophils, is still largely undefined. Using Shp2 knockdown RBL-2H3 (RBL) mast cells, we here reported that Shp2 is required for the activation of RBL cells induced by FcεRI. FcεRΙ-evoked degranulation, calcium mobilization, and synthesis of cytokine transcripts (IL-1β, IL-10, and monocyte chemoattractant protein 1 (MCP-1)) were reduced in Shp2 knockdown RBL cells. Signaling regulatory mechanism investigation using immunoblotting, immunoprecipitation, and GST pull-down assay reveals that the down-regulation of Shp2 expression in RBL cells leads to decreased activities of Fyn, PLCγ, JNK, p38MAPK, and Ras/Erk1/2 after FcεRΙ aggregation. Further studies suggest that Paxillin phosphoryaltion was also impaired, but PAG phosphorylation was normal after FcεRΙ stimulation as a consequence of the inhibition of Shp2 expression in RBL cells. Collectively, our data strongly indicate that Shp2 is essential for the activation of RBL cells in response to FcεRΙ aggregation. Shp2 regulates this process through Fyn and Ras with no involvement of PAG. In addition, we identify Paxillin as an indirect substrate of Shp2 in FcεRΙ-initiated signaling of RBL cells.
Collapse
Affiliation(s)
- Xiaoyun Fang
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Yongjiang Lang
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Yuxiong Wang
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Wei Mo
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Huanhuan Wei
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Jianhui Xie
- Gene Research Center, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Min Yu
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
35
|
Abdel-Rahman SM, Preuett BL. Genetic predictors of susceptibility to cutaneous fungal infections: a pilot genome wide association study to refine a candidate gene search. J Dermatol Sci 2012; 67:147-52. [PMID: 22704677 DOI: 10.1016/j.jdermsci.2012.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 04/27/2012] [Accepted: 05/20/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND Trichophyton tonsurans is the foremost fungal pathogen of minority children in the U.S. Despite overwhelming infection rates, it does not appear that this fungus infects children in a non-specific manner. OBJECTIVE This study was designed to identify genes that may predispose or protect a child from T. tonsurans infection. METHODS Children participating in an earlier longitudinal study wherein infection rates could be reliably determined were eligible for inclusion. DNA from a subset (n=40) of these children at the population extremes underwent whole genome genotyping (WGG). Allele frequencies between cases and controls were examined and significant SNPs were used to develop a candidate gene list for which the remainder of the cohort (n=115) were genotyped. Cumulative infection rate was examined by genotype and the ability of selected genotypes to predict the likelihood of infection explored by multivariable analysis. RESULTS 23 genes with a putative mechanistic role in cutaneous infection were selected for evaluation. Of these, 21 demonstrated significant differences in infection rate between genotypes. A risk index assigned to genotypes in the 21 genes accounted for over 60% of the variability observed in infection rate (adjusted r(2)=0.665, p<0.001). Among these, 8 appeared to account for the majority of variability that was observed (r(2)=0.603, p<0.001). These included genes involved in: leukocyte activation and migration, extracellular matrix integrity and remodeling, epidermal maintenance and wound repair, and cutaneous permeability. CONCLUSIONS Applying WGG to individuals at the extremes of phenotype can help to guide the selection of candidate genes in populations of small cohorts where disease etiology is likely polygenic in nature.
Collapse
Affiliation(s)
- Susan M Abdel-Rahman
- Division of Pediatric Clinical Pharmacology and Medical Toxicology, The Children's Mercy Hospital and Clinics, Kansas City, MO 64108, USA.
| | | |
Collapse
|
36
|
Sibilano R, Frossi B, Suzuki R, D'Incà F, Gri G, Piconese S, Colombo MP, Rivera J, Pucillo CE. Modulation of FcεRI-dependent mast cell response by OX40L via Fyn, PI3K, and RhoA. J Allergy Clin Immunol 2012; 130:751-760.e2. [PMID: 22564682 DOI: 10.1016/j.jaci.2012.03.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 03/11/2012] [Accepted: 03/29/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND The interaction of mast cells (MCs) with regulatory T cells through the OX40 ligand (OX40L):OX40 axis downregulates FcεRI-dependent immediate hypersensitivity responses both in vitro and in vivo. Little is known on OX40L-mediated intracellular signaling or on the mechanism by which OX40L engagement suppresses MC degranulation. OBJECTIVE We explored the role of OX40L engagement on IgE/antigen-triggered MCs both in vitro and in vivo. METHODS The soluble form of OX40 molecule was used to selectively trigger OX40L on MCs in vitro and was used to dissect OX40L contribution in an in vivo model of systemic anaphylaxis. RESULTS OX40L:OX40 interaction led to the recruitment of C-terminal src kinase into lipid rafts, causing a preferential suppression of Fyn kinase activity and subsequent reduction in the phosphorylation of Gab2, the phosphatidylinositol 3-OH kinase regulatory subunit p85, and Akt, without affecting the Lyn pathway. Dampening of Fyn kinase activity also inhibited RhoA activation and microtubule nucleation, key regulators of MC degranulation. The in vivo administration of a blocking antibody to OX40L in wild-type mice caused enhanced immediate hypersensitivity, whereas the administration of soluble OX40 to regulatory T-cell-depleted or OX40-deficient mice reduced MC degranulation. CONCLUSIONS The engagement of OX40L selectively suppresses Fyn-initiated signals required for MC degranulation and serves to limit immediate hypersensitivity. Our data suggest that soluble OX40 can restore the aberrant or absent regulatory T-cell activity, revealing a previously unappreciated homeostatic role for OX40L in setting the basal threshold of MC response.
Collapse
Affiliation(s)
- Riccardo Sibilano
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gab adapter proteins as therapeutic targets for hematologic disease. Adv Hematol 2011; 2012:380635. [PMID: 22216034 PMCID: PMC3246295 DOI: 10.1155/2012/380635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 08/30/2011] [Accepted: 09/06/2011] [Indexed: 12/19/2022] Open
Abstract
The Grb-2 associated binder (Gab) family of scaffolding/adaptor/docking proteins is a group of three molecules with significant roles in cytokine receptor signaling. Gabs possess structural motifs for phosphorylation-dependent receptor recruitment, Grb2 binding, and activation of downstream signaling pathways through p85 and SHP-2. In addition, Gabs participate in hematopoiesis and regulation of immune response which can be aberrantly activated in cancer and inflammation. The multifunctionality of Gab adapters might suggest that they would be too difficult to consider as candidates for “targeted” therapy. However, the one drug/one target approach is giving way to the concept of one drug/multiple target approach since few cancers are addicted to a single signaling molecule for survival and combination drug therapies can be problematic. In this paper, we cover recent findings on Gab multi-functionality, binding partners, and their role in hematological malignancy and examine the concept of Gab-targeted therapy.
Collapse
|