1
|
Ignacio A, Cipelli M, Takiishi T, Favero Aguiar C, Fernandes Terra F, Ghirotto B, Martins Silva E, Castoldi A, Magalhães YT, Antonio T, Nunes Padovani B, Ioshie Hiyane M, Andrade-Oliveira V, Forti FL, Olsen Saraiva Camara N. Lack of mTORC2 signaling in CD11c+ myeloid cells inhibits their migration and ameliorates experimental colitis. J Leukoc Biol 2024; 116:779-792. [PMID: 38652699 DOI: 10.1093/jleuko/qiae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/30/2024] [Accepted: 03/05/2024] [Indexed: 04/25/2024] Open
Abstract
The mammalian target of rapamycin (mTOR) pathway plays a key role in determining immune cells function through modulation of their metabolic status. By specific deletion of Rictor in CD11c+ myeloid cells (referred to here as CD11cRicΔ/Δ), we investigated the role of mTOR complex 2 (mTORC2) signaling in dendritic cells (DCs) function in mice. We showed that upon dextran sulfate sodium-induced colitis, the lack of mTORC2 signaling CD11c+ cells diminishes the colitis score and abrogates DC migration to the mesenteric lymph nodes, thereby diminishing the infiltration of T helper 17 cells in the lamina propria and subsequent inflammation. These findings corroborate with the abrogation of cytoskeleton organization and the decreased activation of Rac1 and Cdc42 GTPases observed in CD11c+-mTORC2-deficient cells. Meta-analysis on colonic samples from ulcerative colitis patients revealed increased gene expression of proinflammatory cytokines, which coincided with augmented expression of the mTOR pathway, a positive correlation between the DC marker ITGAX and interleukin-6, the expression of RICTOR, and CDC42. Together, this work proposes that targeting mTORC2 on DCs offers a key to hamper inflammatory responses, and this way, ameliorates the progression and severity of intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Aline Ignacio
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Marcella Cipelli
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Tatiane Takiishi
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Cristhiane Favero Aguiar
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Fernanda Fernandes Terra
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Bruno Ghirotto
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Eloisa Martins Silva
- Center for Natural and Human Sciences, Federal University of ABC. Alameda da Universidade (UFABC) 09606045, São Bernardo do Campo, SP, Brazil
| | - Angela Castoldi
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Yuli Thamires Magalhães
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo. Av. Prof. Lineu Prestes, 748 05508900, São Paulo, Brazil
| | - Tiago Antonio
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Barbara Nunes Padovani
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Meire Ioshie Hiyane
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Vinicius Andrade-Oliveira
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
| | - Fabio Luis Forti
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo. Av. Prof. Lineu Prestes, 748 05508900, São Paulo, Brazil
| | - Niels Olsen Saraiva Camara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo. Av. Prof. Lineu Prestes 1730, ICB IV, 05508000 São Paulo, Brazil
- Laboratory of Renal Physiology, Department of Medicine, Federal University of São Paulo (UNIFESP). Rua Botucatu 740, 04023-062, São Paulo, Brazil
| |
Collapse
|
2
|
Chen Y, Chen Y, Liu W. Serum cell division control 42 reflects treatment response and survival profiles in recurrent or metastatic oral squamous cell carcinoma patients who receive programmed death-1 inhibitors. Int Immunopharmacol 2024; 129:111547. [PMID: 38290205 DOI: 10.1016/j.intimp.2024.111547] [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] [Received: 11/15/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVES Cell division control 42 (CDC42) facilitates tumor growth, migration, and immune escape to accelerate the pathogenesis and progression of oral squamous cell carcinoma (OSCC). This study intended to explore the optimal cut-value of serum CDC42 for predicting treatment response to programmed death-1 (PD-1) inhibitors and survival in recurrent or metastatic (R/M) OSCC patients. METHODS CDC42 was detected from serum by enzyme-linked immunosorbent assay in 45 R/M OSCC patients before initiating PD-1 inhibitors with or without chemotherapy. Different cutoff values (500, 600, 700, and 800 pg/mL) of CDC42 were selected for further analyses. RESULTS The median (interquartile range) value of CDC42 was 604.0 (477.5-867.5) pg/mL in R/M OSCC patients. Generally, objective response rate (ORR) and disease control rate (DCR) were 37.8 % and 62.2 %. Additionally, ORR (P = 0.030) and DCR (P = 0.004) were decreased in patients with CDC42 ≥ 700 pg/mL versus those with CDC42 < 700 pg/mL; meanwhile, DCR was also reduced in patients with CDC42 ≥ 800 pg/mL versus those with CDC42 < 800 pg/mL (P = 0.014). Interestingly, CDC42 ≥ 600 (P = 0.023), 700 (P = 0.007), and 800 (P = 0.039) pg/mL were related to shorter progression-free survival (PFS). While only CDC42 ≥ 700 (P = 0.004) and 800 (P = 0.046) pg/mL were correlated with worse overall survival (OS). After adjustment, only CDC42 ≥ 700 pg/mL (yes vs. no) independently estimated poor PFS (hazard ratio (HR) = 2.637, P = 0.005) and OS (HR = 5.824, P < 0.001). CONCLUSION CDC42 ≥ 700 pg/mL exerts the optimal prognostic ability to reflect poor treatment response and survival profiles in R/M OSCC patients who receive PD-1 inhibitors.
Collapse
Affiliation(s)
- Yangyi Chen
- Department of Oral and Maxillofacial Surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Yongge Chen
- Department of Oncology, Handan Central Hospital, Handan 056001, Hebei Province, China.
| | - Weixian Liu
- Department of Oral and Maxillofacial Surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China.
| |
Collapse
|
3
|
Bozic I, Lavrnja I. Thiamine and benfotiamine: Focus on their therapeutic potential. Heliyon 2023; 9:e21839. [PMID: 38034619 PMCID: PMC10682628 DOI: 10.1016/j.heliyon.2023.e21839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Thiamine, also known as vitamin B1, is an essential nutrient that plays a crucial role in energy metabolism and overall health. It is a water-soluble vitamin that plays an important role in the conversion of carbohydrates into energy in the body. Thiamine is essential for the proper functioning of the nervous system, heart and muscles. Thiamine deficiency is a life-threatening disease that leads to various disorders and lesions in the nerves and brain, at least in vertebrates. Several thiamine precursors with higher bioavailability have been developed to compensate for thiamine deficiency, including benfotiamine. Benfotiamine is more bioavailable and has higher tissue penetration than thiamine. Studies have shown its antioxidant and anti-inflammatory potential in activated immune and glial cells. It also improves complications observed in type 2 diabetes and has beneficial effects in mouse models of neurodegenerative disease. Benfotiamine represents an off-the-shelf agent used to support nerve health, promote healthy aging and support glucose metabolism. Accordingly, the present review aimed to provide an overview of the neuroprotective effects of thiamine/benfotiamine in the context of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
4
|
Kumar R, Pereira RS, Niemann J, Azimpour AI, Zanetti C, Karantanou C, Minka W, Minciacchi VR, Kowarz E, Meister M, Godavarthy PS, Maguer-Satta V, Lefort S, Wiercinska E, Bonig H, Marschalek R, Krause DS. The differential role of the lipid raft-associated protein flotillin 2 for progression of myeloid leukemia. Blood Adv 2022; 6:3611-3624. [PMID: 35298613 PMCID: PMC9631564 DOI: 10.1182/bloodadvances.2021005992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/08/2022] [Indexed: 11/20/2022] Open
Abstract
Lipid raft-associated proteins play a vital role in membrane-mediated processes. The lipid microdomain-associated protein flotillin 2 (FLOT2), which has a scaffolding function, is involved in polarization, as well as in actin cytoskeletal organization of primitive and mature hematopoietic cells and has been associated with different malignancies. However, its involvement in myeloid leukemias is not well studied. Using murine transplantation models, we show here that the absence of FLOT2 from leukemia-initiating cells (LICs) altered the disease course of BCR-ABL1+ chronic myeloid leukemia (CML), but not of MLL-AF9-driven acute myeloid leukemia (AML). While FLOT2 was required for expression of the adhesion molecule CD44 on both CML- and AML-LIC, a defect in the cytoskeleton, cell polarity, and impaired homing ability of LIC was only observed in FLOT2-deficient BCR-ABL1+ compared with MLL-AF9+ cells. Downstream of CD44, BCR-ABL1 kinase-independent discrepancies were observed regarding expression, localization, and activity of cell division control protein 42 homolog (CDC42) between wild-type (WT) and FLOT2-deficient human CML and AML cells. Inhibition of CDC42 by ML141 impaired the homing of CML LIC and, thereby, CML progression. This suggested that alteration of both CD44 and CDC42 may be causative of impaired CML progression in the absence of FLOT2. In summary, our data suggest a FLOT2-CD44-CDC42 axis, which differentially regulates CML vs AML progression, with deficiency of FLOT2 impairing the development of CML.
Collapse
Affiliation(s)
- Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Raquel S. Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Julian Niemann
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Alexander I. Azimpour
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Costanza Zanetti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Christina Karantanou
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Wahyu Minka
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Valentina R. Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Eric Kowarz
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Melanie Meister
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Parimala S. Godavarthy
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | | | - Sylvain Lefort
- CRCL, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Lyon, France
| | - Eliza Wiercinska
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt, Germany
| | - Halvard Bonig
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt, Germany
- Goethe University, Institute for Transfusion Medicine and Immunohematology, Frankfurt, Germany
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Daniela S. Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Germany
- Frankfurt Cancer Institute, Frankfurt, Germany; and
- Institute for General Pharmacology and Toxicology, Institute for Biochemistry II, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
5
|
Gao S, Xue J, Wu X, Zhong T, Zhang Y, Li S. The relation of blood cell division control protein 42 level with disease risk, comorbidity, tumor features/markers, and prognosis in colorectal cancer patients. J Clin Lab Anal 2022; 36:e24572. [PMID: 35735582 PMCID: PMC9279954 DOI: 10.1002/jcla.24572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Cell division control protein 42 (CDC42) is involved in colorectal cancer (CRC) progression by modulating CD8+ T cell activation, immune escape, and direct oncogenetic biological processes. This study aimed to explore the correlation of blood CDC42 with disease risk, comorbidities, disease features, tumor markers, and prognosis among CRC patients. METHODS CDC42 in peripheral blood mononuclear cells was detected by reverse transcription-quantitative polymerase chain reaction from 250 resectable CRC patients and 50 healthy controls (HCs). CDC42 was divided by quartiles, as well as high and low expressions in CRC patients for correlation and survival analysis. RESULTS CDC42 was elevated in CRC patients vs. HCs (p < 0.001), which had a good ability to distinguish CRC patients from HCs with the area under the curve (95% confidence interval) of 0.889 (0.841-0.937). In CRC patients, CDC42 was not associated with demographics or comorbidities (all p > 0.05), while its higher quartile was linked to increased T stage (p < 0.001), N stage (p = 0.009), TNM stage (p < 0.001), abnormal carcinoembryonic antigen (p = 0.043), and adjuvant chemotherapy administration (p = 0.002). Higher CDC42 quartile (p = 0.002) and CDC42 high (vs. low) (p < 0.001) were related to worse disease-free survival (DFS); meanwhile, elevated CDC42 quartile (p = 0.002) and CDC42 high (vs. low) (p = 0.001) were also linked to poor overall survival (OS). Multivariate Cox's regression analysis presented that CDC42 quartile 3 and 4 (vs. quartile 1) independently predicted declined DFS and OS (all p < 0.05). CONCLUSION Circulating CDC42 relates to higher disease risk, T, N, and TNM stage, abnormal tumor marker, and poor prognosis among CRC patients.
Collapse
Affiliation(s)
- Shuquan Gao
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Jun Xue
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Tingting Zhong
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yingchun Zhang
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Shaodong Li
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| |
Collapse
|
6
|
Olivier JF, Fodil N, Al Habyan S, Gopal A, Artusa P, Mandl JN, McCaffrey L, Gros P. CCDC88B is required for mobility and inflammatory functions of dendritic cells. J Leukoc Biol 2020; 108:1787-1802. [PMID: 32480428 DOI: 10.1002/jlb.3a0420-386r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 04/13/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
The Coiled Coil Domain Containing Protein 88B (CCDC88B) gene is associated with susceptibility to several inflammatory diseases in humans and its inactivation in mice protects against acute neuroinflammation and models of intestinal colitis. We report that mice lacking functional CCDC88B (Ccdc88bMut ) are defective in several dendritic cells (DCs)-dependent inflammatory and immune reactions in vivo. In these mice, an inflammatory stimulus (LPS) fails to induce the recruitment of DCs into the draining lymph nodes (LNs). In addition, OVA-pulsed Ccdc88bMut DCs injected in the footpad do not induce recruitment and activation of antigen-specific CD4+ and CD8+ T cells in their draining LN. Experiments in vitro indicate that this defect is independent of the ability of mutant DCs to capture and present peptide antigen to T cells. Rather, kinetic analyses in vivo of wild-type and Ccdc88bMut DCs indicate a reduced migration capacity in the absence of the CCDC88B protein expression. Moreover, using time-lapse light microscopy imaging, we show that Ccdc88bMut DCs have an intrinsic motility defect. Furthermore, in vivo studies reveal that these reduced migratory properties lead to dampened contact hypersensitivity reactions in Ccdc88b mutant mice. These findings establish a critical role of CCDC88B in regulating movement and migration of DCs. Thus, regulatory variants impacting Ccdc88b expression in myeloid cells may cause variable degrees of DC-dependent inflammatory response in situ, providing a rationale for the genetic association of CCDC88B with several inflammatory and autoimmune diseases in humans.
Collapse
Affiliation(s)
- Jean-Frederic Olivier
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Nassima Fodil
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Sara Al Habyan
- Department of Oncology, McGill University, Montreal, Quebec, Canada.,Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Angelica Gopal
- McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada.,Department of Chemistry, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Patricio Artusa
- McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Judith N Mandl
- McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Luke McCaffrey
- Department of Oncology, McGill University, Montreal, Quebec, Canada.,Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Kishimoto T, Tomishige N, Murate M, Ishitsuka R, Schaller H, Mély Y, Ueda K, Kobayashi T. Cholesterol asymmetry at the tip of filopodia during cell adhesion. FASEB J 2020; 34:6185-6197. [PMID: 32162745 DOI: 10.1096/fj.201900065rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/28/2022]
Abstract
During adhesion, cells develop filopodia to facilitate the attachment to the extracellular matrix. The small guanosine triphosphate (GTP)-binding protein, Cdc42, plays a central role in the formation of filopodia. It has been reported that Cdc42 activity is regulated by cholesterol (Chol). We examined Chol distribution in filopodia using Chol-binding domain 4 (D4) fragment of bacterial toxin, perfringolysin O that senses high membrane concentration of Chol. Our results indicate that fluorescent D4 was enriched at the tip of the outer leaflet of filopodia in the initiation phase of cell adhesion. This enrichment was accompanied by a defect of D4 labeling in the inner leaflet. Steady phase adhered cell experiment indicated that both Cdc42 and ATP-binding cassette transporter, ABCA1, were involved in the binding of D4 to the cell surface. Depletion of Chol activated Cdc42. Our results suggest that asymmetric distribution of Chol at the tip of filopodia induces activation of Cdc42, and thus, facilitates filopodia formation.
Collapse
Affiliation(s)
- Takuma Kishimoto
- Lipid Biology Laboratory, RIKEN, Saitama, Japan.,Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Life Science, Sapporo, Japan
| | - Nario Tomishige
- Lipid Biology Laboratory, RIKEN, Saitama, Japan.,UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | - Motohide Murate
- Lipid Biology Laboratory, RIKEN, Saitama, Japan.,UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | | | - Hubert Schaller
- Institut de Biologie Moléculaire des Plantes, UPR 2357, CNRS, Université de Strasbourg, Strasbourg, France
| | - Yves Mély
- UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | - Kazumitsu Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, Saitama, Japan.,UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| |
Collapse
|
8
|
Zhang Q, Conley SM, Li G, Yuan X, Li PL. Rac1 GTPase Inhibition Blocked Podocyte Injury and Glomerular Sclerosis during Hyperhomocysteinemia via Suppression of Nucleotide-Binding Oligomerization Domain-Like Receptor Containing Pyrin Domain 3 Inflammasome Activation. Kidney Blood Press Res 2019; 44:513-532. [PMID: 31266025 PMCID: PMC6800118 DOI: 10.1159/000500457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Elevated homocysteine (Hcy) levels have been shown to activate nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome leading to podocyte dysfunction and glomerular injury. However, it remains unclear how this inflammasome activation in podocytes is a therapeutic target for reversal of glomerular injury and ultimate sclerosis. The present study tested whether inhibition of Rac1 GTPase activity suppresses NLRP3 inflammation activation and thereby blocks podocyte injury induced by elevated Hcy. In cultured podocytes, we found that L-Hcy (the active Hcy form) stimulated the NLRP3 inflammasome formation, as shown by increased colocalization of NLRP3 with apoptosis-associated speck-like protein (ASC) or caspase-1, which was accompanied by increased interleukin-1β production and caspase-1 activity, indicating NLRP3 inflammasome activation. Rac1 activator, uridine triphosphate (UTP), mimicked L-Hcy-induced NLRP3 inflammasome activation, while Rac1 inhibitor NSC23766 blocked it. This Rac1 inhibition also prevented L-Hcy-induced podocyte dysfunction. All these effects were shown to be mediated via lipid raft redox signaling platforms with nicotinamide adenine dinucleotide phosphate oxidase subunits and consequent O2− production. In animal studies, hyperhomocysteinemia (hHcy) induced by folate-free diet was shown to induce NLRP3 inflammasome formation and activation in glomeruli, which was also mimicked by UTP and inhibited by NSC23766 to a comparable level seen in Nlrp3 gene knockout mice. These results together suggest that Rac1 inhibition protects the kidney from hHcy-induced podocyte injury and glomerular sclerosis due to its action to suppress NLRP3 inflammasome activation in podocytes.
Collapse
Affiliation(s)
- Qinghua Zhang
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sabena M Conley
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Guangbi Li
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Xinxu Yuan
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pin-Lan Li
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA,
| |
Collapse
|
9
|
Keresztes A, Streicher JM. Synergistic interaction of the cannabinoid and death receptor systems - a potential target for future cancer therapies? FEBS Lett 2017; 591:3235-3251. [PMID: 28948607 DOI: 10.1002/1873-3468.12863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/12/2017] [Accepted: 09/19/2017] [Indexed: 01/16/2023]
Abstract
Cannabinoid receptors have been shown to interact with other receptors, including tumor necrosis factor receptor superfamily (TNFRS) members, to induce cancer cell death. When cannabinoids and death-inducing ligands (including TNF-related apoptosis-inducing ligand) are administered together, they have been shown to synergize and demonstrate enhanced antitumor activity in vitro. Certain cannabinoid ligands have been shown to sensitize cancer cells and synergistically interact with members of the TNFRS, thus suggesting that the combination of cannabinoids with death receptor (DR) ligands induces additive or synergistic tumor cell death. This review summarizes recent findings on the interaction of the cannabinoid and DR systems and suggests possible clinical co-application of cannabinoids and DR ligands in the treatment of various malignancies.
Collapse
Affiliation(s)
- Attila Keresztes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - John M Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
10
|
Schulz AM, Stutte S, Hogl S, Luckashenak N, Dudziak D, Leroy C, Forné I, Imhof A, Müller SA, Brakebusch CH, Lichtenthaler SF, Brocker T. Cdc42-dependent actin dynamics controls maturation and secretory activity of dendritic cells. J Cell Biol 2016; 211:553-67. [PMID: 26553928 PMCID: PMC4639873 DOI: 10.1083/jcb.201503128] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cdc42 control of actin dynamics keeps DCs in an immature state, and loss of Cdc42 activity facilitates secretion and rapid up-regulation of intracellular molecules to the cell surface, which shows that Cdc42 contributes to DC immunogenicity by regulating the DC actin cytoskeleton. Cell division cycle 42 (Cdc42) is a member of the Rho guanosine triphosphatase family and has pivotal functions in actin organization, cell migration, and proliferation. To further study the molecular mechanisms of dendritic cell (DC) regulation by Cdc42, we used Cdc42-deficient DCs. Cdc42 deficiency renders DCs phenotypically mature as they up-regulate the co-stimulatory molecule CD86 from intracellular storages to the cell surface. Cdc42 knockout DCs also accumulate high amounts of invariant chain–major histocompatibility complex (MHC) class II complexes at the cell surface, which cannot efficiently present peptide antigens (Ag’s) for priming of Ag-specific CD4 T cells. Proteome analyses showed a significant reduction in lysosomal MHC class II–processing proteins, such as cathepsins, which are lost from DCs by enhanced secretion. As these effects on DCs can be mimicked by chemical actin disruption, our results propose that Cdc42 control of actin dynamics keeps DCs in an immature state, and cessation of Cdc42 activity during DC maturation facilitates secretion as well as rapid up-regulation of intracellular molecules to the cell surface.
Collapse
Affiliation(s)
- Anna M Schulz
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Susanne Stutte
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Sebastian Hogl
- Deutsches Zentrum für Neurodegenerative Erkrankungen, 81377 Munich, Germany
| | - Nancy Luckashenak
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Diana Dudziak
- Department of Dermatology, University Hospital of Erlangen, 91052 Erlangen, Germany
| | - Céline Leroy
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Ignasi Forné
- Adolf Butenandt Institute, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Axel Imhof
- Adolf Butenandt Institute, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Stephan A Müller
- Deutsches Zentrum für Neurodegenerative Erkrankungen, 81377 Munich, Germany
| | - Cord H Brakebusch
- Molecular Pathology Section, Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stefan F Lichtenthaler
- Munich Cluster for Systems Neurology, Ludwig Maximilians University Munich, 80336 Munich, Germany Deutsches Zentrum für Neurodegenerative Erkrankungen, 81377 Munich, Germany Neuroproteomics, Klinikum rechts der Isar, Institute for Advanced Study, Technische Universität München, 80333 Munich, Germany
| | - Thomas Brocker
- Institute for Immunology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| |
Collapse
|
11
|
Brachet A, Norwood S, Brouwers JF, Palomer E, Helms JB, Dotti CG, Esteban JA. LTP-triggered cholesterol redistribution activates Cdc42 and drives AMPA receptor synaptic delivery. ACTA ACUST UNITED AC 2015; 208:791-806. [PMID: 25753037 PMCID: PMC4362467 DOI: 10.1083/jcb.201407122] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cholesterol acts as a sensor of NMDA receptor activation and as a trigger of downstream signaling by engaging small GTPase activation and AMPA receptor synaptic delivery during long-term potentiation. Neurotransmitter receptor trafficking during synaptic plasticity requires the concerted action of multiple signaling pathways and the protein transport machinery. However, little is known about the contribution of lipid metabolism during these processes. In this paper, we addressed the question of the role of cholesterol in synaptic changes during long-term potentiation (LTP). We found that N-methyl-d-aspartate–type glutamate receptor (NMDAR) activation during LTP induction leads to a rapid and sustained loss or redistribution of intracellular cholesterol in the neuron. A reduction in cholesterol, in turn, leads to the activation of Cdc42 and the mobilization of GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid–type glutamate receptors (AMPARs) from Rab11-recycling endosomes into the synaptic membrane, leading to synaptic potentiation. This process is accompanied by an increase of NMDAR function and an enhancement of LTP. These results imply that cholesterol acts as a sensor of NMDAR activation and as a trigger of downstream signaling to engage small GTPase (guanosine triphosphatase) activation and AMPAR synaptic delivery during LTP.
Collapse
Affiliation(s)
- Anna Brachet
- Departamento de Neurobiología, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Stephanie Norwood
- Departamento de Neurobiología, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Jos F Brouwers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, 3508 Utrecht, Netherlands
| | - Ernest Palomer
- Departamento de Neurobiología, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - J Bernd Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, 3508 Utrecht, Netherlands
| | - Carlos G Dotti
- Departamento de Neurobiología, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - José A Esteban
- Departamento de Neurobiología, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
12
|
Holst K, Guseva D, Schindler S, Sixt M, Braun A, Chopra H, Pabst O, Ponimaskin E. Serotonin receptor 5-HT7 regulates morphology and migratory properties of dendritic cells. J Cell Sci 2015; 128:2866-80. [DOI: 10.1242/jcs.167999] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells endowed with the unique ability to initiate adaptive immune responses upon inflammation. Inflammatory processes are often associated with an increased production of serotonin, which operates by activating specific receptors. However, the functional role of serotonin receptors in regulation of DC functions is poorly understood. Here we demonstrate that expression of serotonin receptor 5-HT7 (5-HT7R) as well as its down-stream effector Cdc42 is upregulated in DCs upon maturation. While DC maturation was independent of 5-HT7R, receptor stimulation affected DC morphology via Cdc42-mediated signaling. In addition, basal activity of 5-HT7R was required for the proper expression of the chemokine receptor CCR7, which is a key factor to control DC migration. Consistently, we observed that 5-HT7R enhances chemotactic motility of DCs in vitro by modulating their directionality and migration velocity. Accordingly, migration of DCs in murine colon explants was abolished after pharmacological receptor blockade. Our results indicate a critical role of 5-HT7R/Cdc42-mediated signaling in regulation of DC morphology and motility, suggesting 5-HT7R as a novel target for treatment of a variety of inflammatory and immune disorders.
Collapse
Affiliation(s)
- Katrin Holst
- Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
| | - Daria Guseva
- Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
| | - Susann Schindler
- Department of Airway Immunology, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - Michael Sixt
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Armin Braun
- Department of Airway Immunology, Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - Himpriya Chopra
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Oliver Pabst
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
- Institute of Molecular Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
13
|
Bisel B, Calamai M, Vanzi F, Pavone FS. Decoupling polarization of the Golgi apparatus and GM1 in the plasma membrane. PLoS One 2013; 8:e80446. [PMID: 24312472 PMCID: PMC3846482 DOI: 10.1371/journal.pone.0080446] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 10/03/2013] [Indexed: 01/19/2023] Open
Abstract
Cell polarization is a process of coordinated cellular rearrangements that prepare the cell for migration. GM1 is synthesized in the Golgi apparatus and localized in membrane microdomains that appear at the leading edge of polarized cells, but the mechanism by which GM1 accumulates asymmetrically is unknown. The Golgi apparatus itself becomes oriented toward the leading edge during cell polarization, which is thought to contribute to plasma membrane asymmetry. Using quantitative image analysis techniques, we measure the extent of polarization of the Golgi apparatus and GM1 in the plasma membrane simultaneously in individual cells subject to a wound assay. We find that GM1 polarization starts just 10 min after stimulation with growth factors, while Golgi apparatus polarization takes 30 min. Drugs that block Golgi polarization or function have no effect on GM1 polarization, and, conversely, inhibiting GM1 polarization does not affect Golgi apparatus polarization. Evaluation of Golgi apparatus and GM1 polarization in single cells reveals no correlation between the two events. Our results indicate that Golgi apparatus and GM1 polarization are controlled by distinct intracellular cascades involving the Ras/Raf/MEK/ERK and the PI3K/Akt/mTOR pathways, respectively. Analysis of cell migration and invasion suggest that MEK/ERK activation is crucial for two dimensional migration, while PI3K activation drives three dimensional invasion, and no cumulative effect is observed from blocking both simultaneously. The independent biochemical control of GM1 polarity by PI3K and Golgi apparatus polarity by MEK/ERK may act synergistically to regulate and reinforce directional selection in cell migration.
Collapse
Affiliation(s)
- Blaine Bisel
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- * E-mail:
| | - Martino Calamai
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- National Institute of Optics, National Research Council of Italy (CNR), Florence, Italy
| | - Francesco Vanzi
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
- Department of Evolutionary Biology “Leo Pardi”, University of Florence, Florence, Italy
| | - Francesco Saverio Pavone
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
14
|
Geörg M, Maudsdotter L, Tavares R, Jonsson AB. Meningococcal resistance to antimicrobial peptides is mediated by bacterial adhesion and host cell RhoA and Cdc42 signalling. Cell Microbiol 2013; 15:1938-54. [PMID: 23834289 DOI: 10.1111/cmi.12163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 06/06/2013] [Accepted: 06/28/2013] [Indexed: 11/28/2022]
Abstract
Antimicrobial peptides (AMPs) constitute an essential part of the innate immune defence. Pathogenic bacteria have evolved numerous strategies to withstand AMP-mediated killing. The influence of host epithelia on bacterial AMP resistance is, however, still largely unknown. We found that adhesion to pharyngeal epithelial cells protected Neisseria meningitidis, a leading cause of meningitis and sepsis, from the human cathelicidin LL-37, the cationic model amphipathic peptide (MAP) and the peptaibol alamethicin, but not from polymyxin B. Adhesion to primary airway epithelia resulted in a similar increase in LL-37 resistance. The inhibition of selective host cell signalling mediated by RhoA and Cdc42 was found to abolish the adhesion-induced LL-37 resistance by a mechanism unrelated to the actin cytoskeleton. Moreover, N. meningitidis triggered the formation of cholesterol-rich membrane microdomains in pharyngeal epithelial cells, and host cell cholesterol proved to be essential for adhesion-induced resistance. Our data highlight the importance of Rho GTPase-dependent host cell signalling for meningococcal AMP resistance. These results indicate that N. meningitidis selectively exploits the epithelial microenvironment in order to protect itself from LL-37.
Collapse
Affiliation(s)
- Miriam Geörg
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | | | | |
Collapse
|
15
|
Tugizov SM, Herrera R, Palefsky JM. Epstein-Barr virus transcytosis through polarized oral epithelial cells. J Virol 2013; 87:8179-8194. [PMID: 23698302 PMCID: PMC3700193 DOI: 10.1128/jvi.00443-13] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 04/29/2013] [Indexed: 01/28/2023] Open
Abstract
Although Epstein-Barr virus (EBV) is an orally transmitted virus, viral transmission through the oropharyngeal mucosal epithelium is not well understood. In this study, we investigated how EBV traverses polarized human oral epithelial cells without causing productive infection. We found that EBV may be transcytosed through oral epithelial cells bidirectionally, from both the apical to the basolateral membranes and the basolateral to the apical membranes. Apical to basolateral EBV transcytosis was substantially reduced by amiloride, an inhibitor of macropinocytosis. Electron microscopy showed that virions were surrounded by apical surface protrusions and that virus was present in subapical vesicles. Inactivation of signaling molecules critical for macropinocytosis, including phosphatidylinositol 3-kinases, myosin light-chain kinase, Ras-related C3 botulinum toxin substrate 1, p21-activated kinase 1, ADP-ribosylation factor 6, and cell division control protein 42 homolog, led to significant reduction in EBV apical to basolateral transcytosis. In contrast, basolateral to apical EBV transcytosis was substantially reduced by nystatin, an inhibitor of caveolin-mediated virus entry. Caveolae were detected in the basolateral membranes of polarized human oral epithelial cells, and virions were detected in caveosome-like endosomes. Methyl β-cyclodextrin, an inhibitor of caveola formation, reduced EBV basolateral entry. EBV virions transcytosed in either direction were able to infect B lymphocytes. Together, these data show that EBV transmigrates across oral epithelial cells by (i) apical to basolateral transcytosis, potentially contributing to initial EBV penetration that leads to systemic infection, and (ii) basolateral to apical transcytosis, which may enable EBV secretion into saliva in EBV-infected individuals.
Collapse
Affiliation(s)
- Sharof M Tugizov
- Department of Medicine, University of California San Francisco, San Francisco, California, USA.
| | | | | |
Collapse
|
16
|
Luckashenak N, Wähe A, Breit K, Brakebusch C, Brocker T. Rho-family GTPase Cdc42 controls migration of Langerhans cells in vivo. THE JOURNAL OF IMMUNOLOGY 2012; 190:27-35. [PMID: 23209325 DOI: 10.4049/jimmunol.1201082] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Epidermal Langerhans cells (LCs) of the skin represent the prototype migratory dendritic cell (DC) subtype. In the skin, they take up Ag, migrate to the draining lymph nodes, and contribute to Ag transport and immunity. Different depletion models for LCs have revealed contrasting roles and contributions of this cell type. To target the migratory properties of DCs, we generated mice lacking the Rho-family GTPase Cdc42 specifically in DCs. In these animals, the initial seeding of the epidermis with LCs is functional, resulting in slightly reduced Langerhans cell numbers. However, Cdc42-deficient LCs fail to leave the skin in steady state as well as upon stimulation, as they do not enter the skin-draining afferent lymph vessels. Similarly, also other Cdc42-deficient migratory DC subsets fail to home properly to the corresponding draining lymph nodes. We used this novel mouse model, in which LCs are locked out, to demonstrate that these cells contribute substantially to priming of Ag-specific CD4 and CD8 T cell responses upon epicutaneous immunization, but could not detect a role in the induction of contact hypersensitivity to various doses of hapten.
Collapse
Affiliation(s)
- Nancy Luckashenak
- Institute for Immunology, Ludwig-Maximilians-University, D-80336 Munich, Germany
| | | | | | | | | |
Collapse
|
17
|
Xie C, Li N, Chen ZJ, Li BL, Song BL. The small GTPase Cdc42 interacts with Niemann-Pick C1-like 1 (NPC1L1) and controls its movement from endocytic recycling compartment to plasma membrane in a cholesterol-dependent manner. J Biol Chem 2011; 286:35933-35942. [PMID: 21844200 DOI: 10.1074/jbc.m111.270199] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Niemann-Pick C1-like 1 (NPC1L1) is a multi-transmembrane protein that mediates the absorption of dietary and biliary cholesterol through vesicular endocytosis. The subcellular localization of NPC1L1 is regulated by cholesterol. Cholesterol depletion induces the transport of NPC1L1 to plasma membrane (PM) from endocytic recycling compartment that requires MyoVb·Rab11a·Rab11-FIP2 triple complex, and cholesterol-replenishment renders the internalization of NPC1L1 together with cholesterol. Here, we find that GTP-bound Cdc42 interacts with NPC1L1. Cholesterol depletion regulates the activation of Cdc42 and enhances NPC1L1-Cdc42 interaction. Overexpression of constitutive GTP-bound Cdc42 mutant form or knockdown of Cdc42 inhibits the transport of NPC1L1 to the PM and disturbs the cholesterol-regulated binding of NPC1L1 to Rab11a, MyoVb, and actin. Knockdown of Cdc42 downstream effectors N-WASP or Arp3 also leads to the similar results. In liver-specific Cdc42 knock-out (Cdc42 LKO) mice, NPC1L1 fails to localize to bile canaliculi, and the biliary cholesterol cannot be efficiently reabsorbed. These results indicate that Cdc42 controls the cholesterol-regulated transport and localization of NPC1L1, and plays a role in cholesterol absorption.
Collapse
Affiliation(s)
- Chang Xie
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Na Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Zheng-Jun Chen
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bo-Liang Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bao-Liang Song
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China.
| |
Collapse
|
18
|
Jin S, Zhou F, Katirai F, Li PL. Lipid raft redox signaling: molecular mechanisms in health and disease. Antioxid Redox Signal 2011; 15:1043-83. [PMID: 21294649 PMCID: PMC3135227 DOI: 10.1089/ars.2010.3619] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lipid rafts, the sphingolipid and cholesterol-enriched membrane microdomains, are able to form different membrane macrodomains or platforms upon stimulations, including redox signaling platforms, which serve as a critical signaling mechanism to mediate or regulate cellular activities or functions. In particular, this raft platform formation provides an important driving force for the assembling of NADPH oxidase subunits and the recruitment of other related receptors, effectors, and regulatory components, resulting, in turn, in the activation of NADPH oxidase and downstream redox regulation of cell functions. This comprehensive review attempts to summarize all basic and advanced information about the formation, regulation, and functions of lipid raft redox signaling platforms as well as their physiological and pathophysiological relevance. Several molecular mechanisms involving the formation of lipid raft redox signaling platforms and the related therapeutic strategies targeting them are discussed. It is hoped that all information and thoughts included in this review could provide more comprehensive insights into the understanding of lipid raft redox signaling, in particular, of their molecular mechanisms, spatial-temporal regulations, and physiological, pathophysiological relevances to human health and diseases.
Collapse
Affiliation(s)
- Si Jin
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | | | | | | |
Collapse
|
19
|
Hansen SJ, Rushton J, Dekonenko A, Chand HS, Olson GK, Hutt JA, Pickup D, Lyons CR, Lipscomb MF. Cowpox virus inhibits human dendritic cell immune function by nonlethal, nonproductive infection. Virology 2011; 412:411-25. [PMID: 21334039 DOI: 10.1016/j.virol.2011.01.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 11/03/2010] [Accepted: 01/18/2011] [Indexed: 01/14/2023]
Abstract
Orthopoxviruses encode multiple proteins that modulate host immune responses. We determined whether cowpox virus (CPXV), a representative orthopoxvirus, modulated innate and acquired immune functions of human primary myeloid DCs and plasmacytoid DCs and monocyte-derived DCs (MDDCs). A CPXV infection of DCs at a multiplicity of infection of 10 was nonproductive, altered cellular morphology, and failed to reduce cell viability. A CPXV infection of DCs did not stimulate cytokine or chemokine secretion directly, but suppressed toll-like receptor (TLR) agonist-induced cytokine secretion and a DC-stimulated mixed leukocyte reaction (MLR). LPS-stimulated NF-κB nuclear translocation and host cytokine gene transcription were suppressed in CPXV-infected MDDCs. Early viral immunomodulatory genes were upregulated in MDDCs, consistent with early DC immunosuppression via synthesis of intracellular viral proteins. We conclude that a nonproductive CPXV infection suppressed DC immune function by synthesizing early intracellular viral proteins that suppressed DC signaling pathways.
Collapse
Affiliation(s)
- Spencer J Hansen
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Membrane rafts (MRs) are specialized microdomains in the cell membrane with an altered lipid composition. Upon various stimulations, MRs can be clustered to aggregate or recruit NADPH oxidase sub-units and related proteins to form MR redox signalosomes in the membrane of cells like vascular endothelial cells (ECs). Multiple protein complexes, like MR redox signalosomes, are now considered to play a crucial role in the regulation of cell function and in the development of different cell dysfunctions. To form such redox signalosomes, ceramide will be generated from the hydrolysis of sphingomyelin by lysosomal acid sphingomyelinase that has been translocated via lysosome fusion to the MR area. In this brief review, current information is provided to help understand the occurrence and function of MR redox signalosomes. This may increase enthusiasm of the scientific community for further studies on the molecular mechanisms and the functional significance of forming such MR redox signalosomes.
Collapse
Affiliation(s)
- Chun Zhang
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | |
Collapse
|
21
|
Melendez J, Grogg M, Zheng Y. Signaling role of Cdc42 in regulating mammalian physiology. J Biol Chem 2010; 286:2375-81. [PMID: 21115489 DOI: 10.1074/jbc.r110.200329] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cdc42 is a member of the Rho GTPase family of intracellular molecular switches regulating multiple signaling pathways involved in actomyosin organization and cell proliferation. Knowledge of its signaling function in mammalian cells came mostly from studies using the dominant-negative or constitutively active mutant overexpression approach in the past 2 decades. Such an approach imposes a number of experimental limitations related to specificity, dosage, and/or clonal variability. Recent studies by conditional gene targeting of cdc42 in mice have revealed its tissue- and cell type-specific role and provide definitive information of the physiological signaling functions of Cdc42 in vivo.
Collapse
Affiliation(s)
- Jaime Melendez
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, Ohio 45229, USA
| | | | | |
Collapse
|
22
|
Aldinucci A, Rizzetto L, Pieri L, Nosi D, Romagnoli P, Biagioli T, Mazzanti B, Saccardi R, Beltrame L, Massacesi L, Cavalieri D, Ballerini C. Inhibition of Immune Synapse by Altered Dendritic Cell Actin Distribution: A New Pathway of Mesenchymal Stem Cell Immune Regulation. THE JOURNAL OF IMMUNOLOGY 2010; 185:5102-10. [DOI: 10.4049/jimmunol.1001332] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Sitrin RG, Sassanella TM, Landers JJ, Petty HR. Migrating human neutrophils exhibit dynamic spatiotemporal variation in membrane lipid organization. Am J Respir Cell Mol Biol 2009; 43:498-506. [PMID: 19933376 DOI: 10.1165/rcmb.2009-0286oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Highly ordered sphingolipid-enriched lipid raft microdomains (LRMs) within plasma membranes purportedly function as specialized signaling platforms. Leukocyte migration is believed to entail LRM redistribution, but progress in studying LRMs in situ during cell movement has been limited. By using an improved method for imaging the spectral shift of the environmentally sensitive probe, laurdan (expressed as a generalized polarization function), the plasma membrane order (i.e., tight packing of membrane bilayer lipids) of human polymorphonuclear neutrophils (PMNs) was mapped in real time during migration. Morphologically polarized PMNs exhibited prominent LRM clusters at the uropod, where in every instance membrane order was found to oscillate with mean periodicities of 37.0 ± 1.46 and 149.9 ± 9.0 seconds (P < 0.01). LRM aggregates were also demonstrated in punctate and clustered distributions of nonpolarized cells and transiently at the lamellipodia of polarized PMNs. Cellular polarization was not accompanied by an overall increase in membrane order. LRM disorganization with methyl-β-cyclodextrin had small negative effects on cell velocity, but it abrogated directionally biased migration toward chemotactic gradients of FMLP or leukotriene B(4). LRMs disruption also caused redistribution of Rac 1/2 GTPase and GM3 ganglioside away from the lamellipodium, as well as extension of multiple pseudopods simultaneously or in rapid succession, rather than formation of a defined leading edge. Thus, we demonstrate that the plasma membrane order of migrating PMNs changes dynamically, with prominent oscillations consistently seen at the uropod. These findings solidify the existence of rapidly reorganizing LRMs in situ and support a role for LRMs in chemotaxin responsiveness.
Collapse
Affiliation(s)
- Robert G Sitrin
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | | | |
Collapse
|
24
|
Delaguillaumie A, Marin-Esteban V, Setterblad N, Leh L, Assier E, Gelin C, Charron D, Galy A, Mooney N. Contrasting cytoskeletal regulation of MHC class II peptide presentation by human B cells or dendritic cells. Eur J Immunol 2008; 38:1096-105. [DOI: 10.1002/eji.200737455] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
25
|
Tourkova IL, Shurin GV, Wei S, Shurin MR. Small rho GTPases mediate tumor-induced inhibition of endocytic activity of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:7787-93. [PMID: 17548616 DOI: 10.4049/jimmunol.178.12.7787] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The generation, maturation, and function of dendritic cells (DC) have been shown to be markedly compromised in the tumor microenvironment in animals and humans. However, the molecular mechanisms and intracellular pathways involved in the regulation of the DC system in cancer are not yet fully understood. Recently, we have reported on the role of the small Rho GTPase family members Cdc42, Rac1, and RhoA in regulating DC adherence, motility, and Ag presentation. To investigate involvement of small Rho GTPases in dysregulation of DC function by tumors, we next evaluated how Cdc42, Rac1, and RhoA regulated endocytic activity of DC in the tumor microenvironment. We revealed a decreased uptake of dextran 40 and polystyrene beads by DC generated in the presence of different tumor cell lines, including RM1 prostate, MC38 colon, 3LL lung, and B7E3 oral squamous cell carcinomas in vitro and by DC prepared from tumor-bearing mice ex vivo. Impaired endocytic activity of DC cocultured with tumor cells was associated with decreased levels of active Cdc42 and Rac1. Transduction of DC with the dominant negative Cdc42 and Rac1 genes also led to reduced phagocytosis and receptor-mediated endocytosis. Furthermore, transduction of DC with the constitutively active Cdc42 and Rac1 genes restored endocytic activity of DC that was inhibited by the tumors. Thus, our results suggest that tumor-induced dysregulation of endocytic activity of DC is mediated by reduced activity of several members of the small Rho GTPase family, which might serve as new targets for improving the efficacy of DC vaccines.
Collapse
Affiliation(s)
- Irina L Tourkova
- Department of Pathology, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
26
|
Hacquard-Bouder C, Chimenti MS, Giquel B, Donnadieu E, Fert I, Schmitt A, André C, Breban M. Alteration of antigen-independent immunologic synapse formation between dendritic cells from HLA-B27-transgenic rats and CD4+ T cells: selective impairment of costimulatory molecule engagement by mature HLA-B27. ACTA ACUST UNITED AC 2007; 56:1478-89. [PMID: 17469106 DOI: 10.1002/art.22572] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To investigate the molecular mechanism responsible for the reduced capacity of dendritic cells (DCs) from HLA-B27-transgenic rats to form conjugates with naive T cells. METHODS We monitored interactions between DCs derived from HLA-B27-transgenic, HLA-B7-transgenic control, and nontransgenic rats and naive CD4+ T cells. Chemoattraction was studied in Transwell assays, and the formation of an immunologic synapse was examined by videomicroscopy and electron microscopy. Involvement of specific molecules in the defective interaction was examined in antibody-blocking assays. RESULTS T cells migrated normally toward B27 DCs, but upon contact, the frequency of T cells undergoing a Ca2+ response was decreased, indicating impaired immunologic synapse formation. The immunologic synapse formed between B27 DCs and T cells appeared to be normal, as assessed by electron microscopy and by the Ca2+ response. Blocking lymphocyte function-associated antigen 1 on T cells or blocking activated leukocyte cell adhesion molecules on DCs inhibited an equivalent proportion of conjugates from forming between B27 or control DCs and T cells, whereas blocking CD86 on DCs and blocking CD28, CD2, or CD4 on T cells inhibited a greater number of conjugates from forming with control DCs, indicating specific involvement of costimulatory molecules in the reduced formation of conjugates with B27 DCs. Mature B27 molecules on the DC surface were responsible for this decreased formation of conjugates. CONCLUSION In the HLA-B27-transgenic rat model of spondylarthropathy, mature B27 molecules expressed by DCs impair the formation of an antigen-independent immunologic synapse with naive CD4+ T cells by interfering with the engagement of costimulatory molecules. This phenomenon could potentially affect the production and/or maintenance of regulatory T cells and contribute to the expansion of pathogenic CD4+ T cells.
Collapse
Affiliation(s)
- Cécile Hacquard-Bouder
- Institut Cochin, Université René Descartes, CNRS (VMR 8104), Paris, INSERM U 567, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Allen JA, Halverson-Tamboli RA, Rasenick MM. Lipid raft microdomains and neurotransmitter signalling. Nat Rev Neurosci 2006; 8:128-40. [PMID: 17195035 DOI: 10.1038/nrn2059] [Citation(s) in RCA: 652] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lipid rafts are specialized structures on the plasma membrane that have an altered lipid composition as well as links to the cytoskeleton. It has been proposed that these structures are membrane domains in which neurotransmitter signalling might occur through a clustering of receptors and components of receptor-activated signalling cascades. The localization of these proteins in lipid rafts, which is affected by the cytoskeleton, also influences the potency and efficacy of neurotransmitter receptors and transporters. The effect of lipid rafts on neurotransmitter signalling has also been implicated in neurological and psychiatric diseases.
Collapse
Affiliation(s)
- John A Allen
- Department of Physiology and Biophysics, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | | | | |
Collapse
|
28
|
Hunter I, Nixon GF. Spatial compartmentalization of tumor necrosis factor (TNF) receptor 1-dependent signaling pathways in human airway smooth muscle cells. Lipid rafts are essential for TNF-alpha-mediated activation of RhoA but dispensable for the activation of the NF-kappaB and MAPK pathways. J Biol Chem 2006; 281:34705-15. [PMID: 16982613 PMCID: PMC2653078 DOI: 10.1074/jbc.m605738200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor (TNF)-alpha-induced activation of RhoA, mediated by TNF receptor 1 (TNFR1), is a prerequisite step in a pathway that leads to increased 20-kDa light chain of myosin (MLC20) phosphorylation and airway smooth muscle contraction. In this study, we have investigated the proximal events in TNF-alpha-induced RhoA activation. TNFR1 is localized to both lipid raft and nonraft regions of the plasma membrane in primary human airway smooth muscle cells. TNF-alpha engagement of TNFR1 recruited the adaptor proteins TRADD, TRAF-2, and RIP into lipid rafts and activated RhoA, NF-kappaB, and MAPK pathways. Depletion of cholesterol from rafts with methyl-beta-cyclodextrin caused a redistribution of TNFR1 to nonraft plasma membrane and prevented ligand-induced RhoA activation. By contrast, TNF-alpha-induced activation of NF-kappaB and MAPKs was unaffected by methyl-beta-cyclodextrin indicating that, in airway smooth muscle cells, activation of these pathways occurred independently of lipid rafts. Targeted knockdown of caveolin-1 completely abrogated TNF-alpha-induced RhoA activation, identifying this raft-resident protein as a positive regulator of the activation process. The signaling adaptors TRADD and RIP were also found to be necessary for ligand-induced RhoA activation. Taken together, our results suggest that in airway smooth muscle cells, spatial compartmentalization of TNFR1 provides a mechanism for generating distinct signaling outcomes in response to ligand engagement and define a mechanistic role for lipid rafts and caveolin-1 in TNF-alpha-induced activation of RhoA.
Collapse
Affiliation(s)
- Irene Hunter
- School of Medical Sciences, University of Aberdeen, IMS Building, Foresterhill, Aberdeen AB25 2ZD, United Kingdom.
| | | |
Collapse
|
29
|
Désiré L, Bourdin J, Loiseau N, Peillon H, Picard V, De Oliveira C, Bachelot F, Leblond B, Taverne T, Beausoleil E, Lacombe S, Drouin D, Schweighoffer F. RAC1 inhibition targets amyloid precursor protein processing by gamma-secretase and decreases Abeta production in vitro and in vivo. J Biol Chem 2005; 280:37516-25. [PMID: 16150730 DOI: 10.1074/jbc.m507913200] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
beta-Amyloid peptides (Abeta) that form the senile plaques of Alzheimer disease consist mainly of 40- and 42-amino acid (Abeta 40 and Abeta 42) peptides generated from the cleavage of the amyloid precursor protein (APP). Generation of Abeta involves beta-secretase and gamma-secretase activities and is regulated by membrane trafficking of the proteins involved in Abeta production. Here we describe a new small molecule, EHT 1864, which blocks the Rac1 signaling pathways. In vitro, EHT 1864 blocks Abeta 40 and Abeta 42 production but does not impact sAPPalpha levels and does not inhibit beta-secretase. Rather, EHT 1864 modulates APP processing at the level of gamma-secretase to prevent Abeta 40 and Abeta 42 generation. This effect does not result from a direct inhibition of the gamma-secretase activity and is specific for APP cleavage, since EHT 1864 does not affect Notch cleavage. In vivo, EHT 1864 significantly reduces Abeta 40 and Abeta 42 levels in guinea pig brains at a threshold that is compatible with delaying plaque accumulation and/or clearing the existing plaque in brain. EHT 1864 is the first derivative of a new chemical series that consists of candidates for inhibiting Abeta formation in the brain of AD patients. Our findings represent the first pharmacological validation of Rac1 signaling as a target for developing novel therapies for Alzheimer disease.
Collapse
|
30
|
Kriehuber E, Bauer W, Charbonnier AS, Winter D, Amatschek S, Tamandl D, Schweifer N, Stingl G, Maurer D. Balance between NF-κB and JNK/AP-1 activity controls dendritic cell life and death. Blood 2005; 106:175-83. [PMID: 15755895 DOI: 10.1182/blood-2004-08-3072] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The life cycle of dendritic cells (DCs) must be precisely regulated for proper functioning of adaptive immunity. However, signaling pathways actively mediating DC death remain enigmatic. Here we describe a novel mechanism of hierarchical transcriptional control of DC life and death. Ligation of tumor necrosis factor receptor superfamily (TNFR-SF) members on DCs and cognate contact with T cells resulted in quantitatively balanced nuclear factor-κB (NF-κB) and c-Jun N-terminal kinase (JNK)–mediated activator protein-1 (AP-1) induction and strongly enhanced DC longevity. Specific blockade of NF-κB in DCs induced strongly augmented JNK/AP-1 activity because of elevated levels of reactive oxygen species. In this scenario, DC activation by TNFR-SF members or T cells induced DC apoptosis. Specific inhibition of JNK/AP-1 rescued DCs from this activation-induced cell death program and restored TNFR-SF member- and T-cell–mediated survival. We conclude that JNK/AP-1 activity is under negative feedback control of NF-κB and can execute apoptosis in DCs. Thus, feedback-controlled signaling amplitudes of 2 transcriptional pathways decide the fate of a DC.
Collapse
Affiliation(s)
- Ernst Kriehuber
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kuipers HF, Biesta PJ, Groothuis TA, Neefjes JJ, Mommaas AM, van den Elsen PJ. Statins Affect Cell-Surface Expression of Major Histocompatibility Complex Class II Molecules by Disrupting Cholesterol-Containing Microdomains. Hum Immunol 2005; 66:653-65. [PMID: 15993711 DOI: 10.1016/j.humimm.2005.04.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Revised: 04/07/2005] [Accepted: 04/11/2005] [Indexed: 11/18/2022]
Abstract
Statins, the main therapy for hypercholesterolemia, are currently considered as possible immunomodulatory agents. Statins inhibit the production of proinflammatory cytokines and reduce the expression of several immunoregulatory molecules, including major histocompatibility complex class II (MHC-II) molecules. In this study, we investigated the mechanism by which simvastatin reduces the membrane expression of MHC-II molecules on several human cell types. We demonstrate that the reduction of MHC-II membrane expression by simvastatin correlates with disruption of cholesterol-containing microdomains, which transport and concentrate MHC-II molecules to the cell surface. In addition, we demonstrate that statins reduce cell-surface expression of other immunoregulatory molecules, which include MHC-I, CD3, CD4, CD8, CD28, CD40, CD80, CD86, and CD54. Our observations indicate that the downregulation of MHC-II at the cell surface contributes to the immunomodulatory properties of statins and is achieved through disruption of cholesterol-containing microdomains, which are involved in their intracellular transport.
Collapse
Affiliation(s)
- Hedwich F Kuipers
- Division of Molecular Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
32
|
Zeyda M, Säemann MD, Stuhlmeier KM, Mascher DG, Nowotny PN, Zlabinger GJ, Waldhäusl W, Stulnig TM. Polyunsaturated fatty acids block dendritic cell activation and function independently of NF-kappaB activation. J Biol Chem 2005; 280:14293-301. [PMID: 15684433 DOI: 10.1074/jbc.m410000200] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) modulate immune responses leading to clinically significant beneficial effects in a variety of inflammatory disorders. PUFA effects on T cells have been extensively studied, but their influence on human dendritic cells (DCs), which are the most potent antigen-presenting cells and play a key role in initiating immune responses, has not been elucidated so far. Here we show that PUFAs of the n-3 and n-6 series (arachidonic and eicosapentaenoic acid) affect human monocyte-derived DC differentiation and inhibit their activation by LPS, resulting in altered DC surface molecule expression and diminished cytokine secretion. Furthermore, the potency to stimulate T cells was markedly inhibited in PUFA-treated DCs. The PUFA-mediated block in LPS-induced DC activation is reflected by diminished TNF-alpha, IL-12p40, CD40, and COX-2 mRNA levels. Strikingly, typical LPS-induced signaling events such as degradation of IkappaB and activation of NF-kappaB were not affected by PUFAs, even though DC membrane lipid composition was markedly altered. Arachidonic and eicosapentaenoic acid both altered DC prostaglandin production, but inhibitors of cyclooxygenases and lipoxygenases did not abolish PUFA effects, indicating that the observed PUFA actions on DCs were independent of autoregulation via eicosanoids. These data demonstrate a unique interference with DC activation and function that could significantly contribute to the well known anti-inflammatory effects of PUFAs.
Collapse
Affiliation(s)
- Maximilian Zeyda
- Clinical Divisions of Endocrinology and Metabolism, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
For many years after the discovery of actin filaments and microtubules, it was widely assumed that their polymerization, organization, and functions were largely distinct. However, in recent years it has become increasingly apparent that coordinated interactions between microtubules and filamentous actin are involved in many polarized processes, including cell shape, mitotic spindle orientation, motility, growth cone guidance, and wound healing. In the past few years, significant strides have been made in unraveling the intricacies that govern these intertwined cytoskeletal rearrangements.
Collapse
Affiliation(s)
- Atsuko Kodama
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10021, USA
| | | | | |
Collapse
|