1
|
Liu C, Cong X, Wang Y, Guo Q, Xie Y, Geng F, Guo J, Dong L, Zhou Y, Wu H, Yu B, Wu J, Zhang H, Yu X, Kong W. Fast DNA Vaccination Strategy Elicits a Stronger Immune Response Dependent on CD8 +CD11c + Cell Accumulation. Front Oncol 2021; 11:752444. [PMID: 34950581 PMCID: PMC8691261 DOI: 10.3389/fonc.2021.752444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Conventional DNA vaccine strategies usually employ a regimen of immunizations at 2-week or longer intervals to induce effective memory cell-dependent immune responses. Clinical cancer treatment requires a faster immunization strategy to contend with tumor progression. In this study, a novel fast immunization strategy was established, wherein a DNA vaccine was intramuscularly administered on days 0, 2, and 5 in a murine lung cancer model. Effector cells peaked 7 to 10 days after the last vaccination. Compared with traditional 2-week-interval immunization strategies, antigen-specific cytolysis and INF-γ secretion were significantly enhanced under the fast vaccination approach. As a result, the rapidly administered DNA vaccine elicited stronger and more prompt antitumor effects. The probable underlying mechanism of fast immunization was the accumulation of CD8+CD11c+ antigen-presenting cells at the injection site, which enhanced subsequent antigen presentation. In conclusion, the fast DNA vaccination strategy shortened vaccination time to 5 days and elicited a stronger antitumor immune response.
Collapse
Affiliation(s)
- Chenlu Liu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianling Cong
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Yuqian Wang
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Qianqian Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yu Xie
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Fei Geng
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jie Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Ling Dong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yi Zhou
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| |
Collapse
|
2
|
Novoszel P, Holcmann M, Stulnig G, De Sa Fernandes C, Zyulina V, Borek I, Linder M, Bogusch A, Drobits B, Bauer T, Tam‐Amersdorfer C, Brunner PM, Stary G, Bakiri L, Wagner EF, Strobl H, Sibilia M. Psoriatic skin inflammation is promoted by c-Jun/AP-1-dependent CCL2 and IL-23 expression in dendritic cells. EMBO Mol Med 2021; 13:e12409. [PMID: 33724710 PMCID: PMC8033525 DOI: 10.15252/emmm.202012409] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptor (TLR) stimulation induces innate immune responses involved in many inflammatory disorders including psoriasis. Although activation of the AP-1 transcription factor complex is common in TLR signaling, the specific involvement and induced targets remain poorly understood. Here, we investigated the role of c-Jun/AP-1 protein in skin inflammation following TLR7 activation using human psoriatic skin, dendritic cells (DC), and genetically engineered mouse models. We show that c-Jun regulates CCL2 production in DCs leading to impaired recruitment of plasmacytoid DCs to inflamed skin after treatment with the TLR7/8 agonist Imiquimod. Furthermore, deletion of c-Jun in DCs or chemical blockade of JNK/c-Jun signaling ameliorates psoriasis-like skin inflammation by reducing IL-23 production in DCs. Importantly, the control of IL-23 and CCL2 by c-Jun is most pronounced in murine type-2 DCs. CCL2 and IL-23 expression co-localize with c-Jun in type-2/inflammatory DCs in human psoriatic skin and JNK-AP-1 inhibition reduces the expression of these targets in TLR7/8-stimulated human DCs. Therefore, c-Jun/AP-1 is a central driver of TLR7-induced immune responses by DCs and JNK/c-Jun a potential therapeutic target in psoriasis.
Collapse
Affiliation(s)
- Philipp Novoszel
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Martin Holcmann
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Gabriel Stulnig
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Cristiano De Sa Fernandes
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Victoria Zyulina
- Division of Immunology and PathophysiologyOtto Loewi Research CenterMedical University of GrazGrazAustria
| | - Izabela Borek
- Division of Immunology and PathophysiologyOtto Loewi Research CenterMedical University of GrazGrazAustria
| | - Markus Linder
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Alexandra Bogusch
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Barbara Drobits
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Thomas Bauer
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| | - Carmen Tam‐Amersdorfer
- Division of Immunology and PathophysiologyOtto Loewi Research CenterMedical University of GrazGrazAustria
| | - Patrick M Brunner
- Division of Immunology, Allergy and Infectious DiseasesDepartment of DermatologyMedical University of ViennaViennaAustria
| | - Georg Stary
- Division of Immunology, Allergy and Infectious DiseasesDepartment of DermatologyMedical University of ViennaViennaAustria
| | - Latifa Bakiri
- Division of Immunology, Allergy and Infectious DiseasesDepartment of DermatologyMedical University of ViennaViennaAustria
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Erwin F Wagner
- Division of Immunology, Allergy and Infectious DiseasesDepartment of DermatologyMedical University of ViennaViennaAustria
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Herbert Strobl
- Division of Immunology and PathophysiologyOtto Loewi Research CenterMedical University of GrazGrazAustria
| | - Maria Sibilia
- Department of Medicine IComprehensive Cancer CenterInstitute of Cancer ResearchMedical University of ViennaViennaAustria
| |
Collapse
|
3
|
Gao Q, Li F, Wang S, Shen Z, Cheng S, Ping Y, Qin G, Chen X, Yang L, Cao L, Liu S, Zhang B, Wang L, Sun Y, Zhang Y. A cycle involving HMGB1, IFN-γ and dendritic cells plays a putative role in anti-tumor immunity. Cell Immunol 2019; 343:103850. [PMID: 30153900 DOI: 10.1016/j.cellimm.2018.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 07/23/2018] [Accepted: 08/17/2018] [Indexed: 12/30/2022]
|
4
|
Gao Q, Wang S, Chen X, Cheng S, Zhang Z, Li F, Huang L, Yang Y, Zhou B, Yue D, Wang D, Cao L, Maimela NR, Zhang B, Yu J, Wang L, Zhang Y. Cancer-cell-secreted CXCL11 promoted CD8 + T cells infiltration through docetaxel-induced-release of HMGB1 in NSCLC. J Immunother Cancer 2019; 7:42. [PMID: 30744691 PMCID: PMC6371476 DOI: 10.1186/s40425-019-0511-6] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 01/16/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Chemotherapy combined with immunotherapy becomes the main trend in lung cancer intervention; however, how chemotherapy promotes the immune function remains elusive. Therefore, we sought to determine how chemotherapy promotes the immune function. METHODS We determined in 100 NSCLC patients the expression of CD8, functional markers (IFN-γ, Granzyme B, and Perforin) and specific chemokines by quantitative real-time reverse transcriptase-PCR. Functional experiments were carried out to check whether docetaxel (DOC), a chemotherapeutic agent, modifies the expression of HMGB1 and CXCL11, and influences the infiltration properties of CD8+ T cells to the tumor microenvironment. The mechanism of the release of HMGB1 and CXCL11 was determined by flow cytometry, immunofluorescence and western blotting. In in vivo experiment, we confirmed how DOC enhanced the recruitment of HER2-CAR T cells to tumor sites. RESULTS We found that DOC upregulated the expression of chemokine receptor ligand CXCL11 in tumor microenvironment and subsequently enhanced CD8+ T cell recruitment. DOC treatment significantly increased HMGB1 release in an ROS-dependent manner. Recombinant protein HMGB1 stimulated the secretion of CXCL11 via NF-κB activation in vitro. Tumors from DOC-treated mice exhibited higher expression of HMGB1 and CXCL11, more HER2-CAR T cell infiltration, and reduced progression, relative to control. Increased HMGB1 and CXCL11 expressions were positively correlated with prolonged overall survival of lung cancer patients. CONCLUSIONS Our results demonstrate that DOC induces CD8+ T cell recruitment to the tumor microenvironment by enhancing the secretion of HMGB1 and CXCL11, thus improving the anti-tumor efficacy, indicating that modulating the HMGB1-CXCL11 axis might be helpful for NSCLC treatment.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Cell Line, Tumor
- Chemokine CXCL11/immunology
- Chemotaxis, Leukocyte/drug effects
- Docetaxel/pharmacology
- Docetaxel/therapeutic use
- Female
- HMGB1 Protein/genetics
- HMGB1 Protein/immunology
- Humans
- Immunotherapy, Adoptive
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice, Inbred BALB C
- Mice, Nude
- Receptor, ErbB-2/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Qun Gao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shumin Wang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shaoyan Cheng
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Feng Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Lan Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yang Yang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Bin Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China
| | - Dongli Yue
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dan Wang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ling Cao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | | | - Bin Zhang
- Department of Hematology/Oncology, School of Medicine, Northwestern University, Chicago, USA
| | - Jane Yu
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Liping Wang
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, People's Republic of China.
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
5
|
Openshaw RL, Kwon J, McColl A, Penninger JM, Cavanagh J, Pratt JA, Morris BJ. JNK signalling mediates aspects of maternal immune activation: importance of maternal genotype in relation to schizophrenia risk. J Neuroinflammation 2019; 16:18. [PMID: 30691477 PMCID: PMC6350402 DOI: 10.1186/s12974-019-1408-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/11/2019] [Indexed: 12/31/2022] Open
Abstract
Background Important insight into the mechanisms through which gene-environmental interactions cause schizophrenia can be achieved through preclinical studies combining prenatal immune stimuli with disease-related genetic risk modifications. Accumulating evidence associates JNK signalling molecules, including MKK7/MAP2K7, with genetic risk. We tested the hypothesis that Map2k7 gene haploinsufficiency in mice would alter the prenatal immune response to the viral mimetic polyriboinosinic-polyribocytidylic acid (polyI:C), specifically investigating the impact of maternal versus foetal genetic variants. Methods PolyI:C was administered to dams (E12.5), and cytokine/chemokine levels were measured 6 h later, in maternal plasma, placenta and embryonic brain. Results PolyI:C dramatically elevated maternal plasma levels of most cytokines/chemokines. Induction of IL-1β, IL-2, IL-10, IL-12, TNF-α and CXCL3 was enhanced, while CCL5 was suppressed, in Map2k7 hemizygous (Hz) dams relative to controls. Maternal polyI:C administration also increased embryonic brain chemokines, influenced by both maternal and embryonic genotype: CCL5 and CXCL10 levels were higher in embryonic brains from Map2k7 dams versus control dams; for CCL5, this was more pronounced in Map2k7 Hz embryos. Placental CXCL10 and CXCL12 levels were also elevated by polyI:C, the former enhanced and the latter suppressed, in placentae from maternal Map2k7 Hzs relative to control dams receiving polyI:C. Conclusions The results demonstrate JNK signalling as a mediator of MIA effects on the foetus. Since both elevated CXCL10 and supressed CXCL12 compromise developing GABAergic interneurons, the results support maternal immune challenge contributing to schizophrenia-associated neurodevelopmental abnormalities. The influence of Map2k7 on cytokine/chemokine induction converges the genetic and environmental aspects of schizophrenia, and the overt influence of maternal genotype offers an intriguing new insight into modulation of embryonic neurodevelopment by genetic risk. Electronic supplementary material The online version of this article (10.1186/s12974-019-1408-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rebecca L Openshaw
- Institute of Neuroscience and Psychology, West Medical Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Jaedeok Kwon
- Institute of Neuroscience and Psychology, West Medical Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Alison McColl
- Institute of Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Josef M Penninger
- IMBA, Institute for Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Jonathan Cavanagh
- Institute of Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, West Medical Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
6
|
Incrocci R, McAloon J, Montesano M, Bardahl J, Vagvala S, Stone A, Swanson-Mungerson M. Epstein-Barr virus LMP2A utilizes Syk and PI3K to activate NF-κB in B-cell lymphomas to increase MIP-1α production. J Med Virol 2019; 91:845-855. [PMID: 30609049 DOI: 10.1002/jmv.25381] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/02/2018] [Accepted: 12/04/2018] [Indexed: 12/26/2022]
Abstract
The incidence of Hodgkin's lymphoma (HL) is growing due to an increase in Epstein-Barr virus (EBV)-associated HL in AIDS patients. The HL tumor microenvironment is vital for the survival of the malignant Hodgkin-Reed Sternberg (HRS) cells of HL, which express the EBV protein latent membrane protein 2A (LMP2A). While previous work shows that LMP2A mimics B-cell receptor (BCR) signaling to promote the survival of HRS cells, the ability of LMP2A to establish and maintain the tumor microenvironment through the production of chemokines remains unknown. Since BCR signaling induces the production of the chemokine macrophage inflammatory protein-1α (MIP-1α), and since LMP2A is a BCR mimic, we hypothesized that LMP2A increases MIP-1α levels. A comparison of multiple LMP2A-negative and -positive cell lines demonstrates that LMP2A increases MIP-1α. Additionally, LMP2A-mutant cell lines and pharmacologic inhibitors indicate that LMP2A activates a Syk/PI3K/NF-κB pathway to enhance MIP-1α. Finally, based on the finding that an NF-κB inhibitor decreased MIP-1α RNA/protein in LMP2A-positive cells, we are the first to demonstrate that LMP2A increases the nuclear localization of the NF-κB p65 subunit using DNA-binding assays and confocal microscopy in human B cells. These findings not only have implications for the treatment of HL, but also other LMP2A-expressing B-cell tumors that overexpress NF-κB.
Collapse
Affiliation(s)
- Ryan Incrocci
- Department of Microbiology and Immunology, College of Graduate Studies, Midwestern University, Downers Grove, Illinois
| | - Jason McAloon
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois
| | - Michael Montesano
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois.,Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Downers Grove, Illinois
| | - Jonathan Bardahl
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois.,Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Downers Grove, Illinois
| | - Saivenkat Vagvala
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois
| | - Amanda Stone
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois
| | - Michelle Swanson-Mungerson
- Department of Microbiology and Immunology, College of Graduate Studies, Midwestern University, Downers Grove, Illinois.,Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois
| |
Collapse
|
7
|
Jang SY, Hwang J, Kim BS, Lee EY, Oh BH, Kim MH. Structural basis of inactivation of Ras and Rap1 small GTPases by Ras/Rap1-specific endopeptidase from the sepsis-causing pathogen Vibrio vulnificus. J Biol Chem 2018; 293:18110-18122. [PMID: 30282804 PMCID: PMC6254334 DOI: 10.1074/jbc.ra118.004857] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/25/2018] [Indexed: 12/15/2022] Open
Abstract
Multifunctional autoprocessing repeats-in-toxin (MARTX) toxins are secreted by Gram-negative bacteria and function as primary virulence-promoting macromolecules that deliver multiple cytopathic and cytotoxic effector domains into the host cytoplasm. Among these effectors, Ras/Rap1-specific endopeptidase (RRSP) catalyzes the sequence-specific cleavage of the Switch I region of the cellular substrates Ras and Rap1 that are crucial for host innate immune defenses during infection. To dissect the molecular basis underpinning RRSP-mediated substrate inactivation, we determined the crystal structure of an RRSP from the sepsis-causing bacterial pathogen Vibrio vulnificus (VvRRSP). Structural and biochemical analyses revealed that VvRRSP is a metal-independent TIKI family endopeptidase composed of an N-terminal membrane-localization and substrate-recruitment domain (N lobe) connected via an inter-lobe linker to the C-terminal active site-coordinating core β-sheet-containing domain (C lobe). Structure-based mutagenesis identified the 2His/2Glu catalytic residues in the core catalytic domain that are shared with other TIKI family enzymes and that are essential for Ras processing. In vitro KRas cleavage assays disclosed that deleting the N lobe in VvRRSP causes complete loss of enzymatic activity. Endogenous Ras cleavage assays combined with confocal microscopy analysis of HEK293T cells indicated that the N lobe functions both in membrane localization via the first α-helix and in substrate assimilation by altering the functional conformation of the C lobe to facilitate recruitment of cellular substrates. Collectively, these results indicate that RRSP is a critical virulence factor that robustly inactivates Ras and Rap1 and augments the pathogenicity of invading bacteria via the combined effects of its N and C lobes.
Collapse
Affiliation(s)
- Song Yee Jang
- From the Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141,; the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and
| | - Jungwon Hwang
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and.
| | - Byoung Sik Kim
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and; the Department of Food Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Eun-Young Lee
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and
| | - Byung-Ha Oh
- From the Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141,.
| | - Myung Hee Kim
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and.
| |
Collapse
|
8
|
Fischetti L, Zhong Z, Pinder CL, Tregoning JS, Shattock RJ. The synergistic effects of combining TLR ligand based adjuvants on the cytokine response are dependent upon p38/JNK signalling. Cytokine 2017; 99:287-296. [PMID: 28826648 DOI: 10.1016/j.cyto.2017.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/17/2022]
Abstract
Toll like receptor (TLR) ligands are important adjuvant candidates, causing antigen presenting cells to release inflammatory mediators, leading to the recruitment and activation of other leukocytes. The aim of this study was to define the response of human blood derived dendritic cells and macrophages to three TLR ligands acting singly or in combination, Poly I:C (TLR3), GLA (TLR4) and R848 (TLR7/8). Combinations of TLR agonists have been shown to have a synergistic effect on individual cytokines, here we look at the global inflammatory response measuring both cytokines and chemokines. Using a custom Luminex assay we saw dose responses in several mediators including CCL3 (MIP1α), IL-1α, IL-1β, IL-12, CXCL10 (IP-10) and IL-6, all of which were significantly increased by the combination of R848 and GLA, even when low dose GLA was added. The synergistic effect was inhibited by specific MAP kinase inhibitors blocking the kinases p38 and JNK but not MEK1. Combining TLR adjuvants also had a synergistic effect on cytokine responses in human mucosal tissue explants. From this we conclude that the combination of R848 and GLA potentiates the inflammatory profile of antigen presenting cells. Since the pattern of inflammatory mediators released can alter the quality and quantity of the adaptive immune response to vaccination, this study informs vaccine adjuvant design.
Collapse
Affiliation(s)
- Lucia Fischetti
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - Ziyun Zhong
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - Christopher L Pinder
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - John S Tregoning
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - Robin J Shattock
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom.
| |
Collapse
|
9
|
CXCL2/MIF-CXCR2 signaling promotes the recruitment of myeloid-derived suppressor cells and is correlated with prognosis in bladder cancer. Oncogene 2016; 36:2095-2104. [PMID: 27721403 DOI: 10.1038/onc.2016.367] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 08/03/2016] [Accepted: 08/15/2016] [Indexed: 02/07/2023]
Abstract
The accumulation of myeloid-derived suppressor cells (MDSCs) has been observed in solid tumors and is correlated with tumor progression; however, the underlying mechanism is still poorly understood. In this study, we identified a mechanism by which tumor cells induce MDSC accumulation and expansion in the bladder cancer (BC) microenvironment via CXCL2/MIF-CXCR2 signaling. Elevated expression of CXCL2 and MIF and an increased number of CD33+ MDSCs were detected in BC tissues, and these increases were significantly associated with advanced disease stage and poor patient prognosis (P<0.01). A positive association was observed between CXCL2 or MIF expression and the number of tumor-infiltrating CD33+ MDSCs (P<0.01). Subsequently, we demonstrated that CD45+CD33+CD11b+HLA-DR- MDSCs from fresh BC tissues displayed high levels of suppressive molecules, including Arg1, iNOS, ROS, PDL-1 and P-STAT3, and stronger suppression of T-cell proliferation. Interestingly, these CD45+CD33+CD11b+HLA-DR- MDSCs exhibited increased CXCR2 expression compared with that in peripheral blood from BC patients or healthy controls (P<0.05). Chemotaxis assay revealed that bladder cancer cell line J82 induced MDSC migration via CXCL2/MIF-CXCR2 signaling in vitro. Mechanistic studies demonstrated that J82-induced MDSC trafficking and CXCR2 expression were associated with increased phosphorylation of p38, ERK and p65. Conversely, inhibition of the phosphorylation of p38, ERK or p65 decreased J82-induced MDSC trafficking and CXCR2 expression. CXCL2/MIF-stimulated activation of the mitogen-activated protein kinase and nuclear factor kappa B pathways in MDSCs was MyD88 dependent. Overall, our results identify the CXCL2/MIF-CXCR2 axis as an important mediator in MDSC recruitment and as predictors and potential therapeutic targets in BC patients.
Collapse
|
10
|
Zhu M, Xu W, Su H, Huang Q, Wang B. Addition of CpG ODN and Poly (I:C) to a standard maturation cocktail generates monocyte-derived dendritic cells and induces a potent Th1 polarization with migratory capacity. Hum Vaccin Immunother 2016; 11:1596-605. [PMID: 26039883 DOI: 10.1080/21645515.2015.1046659] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Monocyte-derived dendritic cells (DCs) are used as immunoadjuvant cells in cancer vaccines and have made great progress. However, an optimal DCs subset is vital for this treatment effect, the current 'gold standard' cytokine cocktail DCs have a shortcoming in their cytokines secretion, especially IL-12p70, mainly because of the existence of PGE2. Therefore, it is necessary to find an appropriate DCs-based immunotherapeutic protocol. In this study, we compared a novel 'improved' maturation cytokine cocktail with the current 'gold standard' maturation cytokine cocktail used for generating standard DCs. The 'improved' maturation cytokine cocktail DCs showed a higher levels surface markers expression (CD80, CD83, CD86 and HLA-DR), the chemokine receptors CXCR4 and CCR7 and chemokine CCL19, CCL21 and CXCL21, whereas CCR5 expression was reduced. Most importantly, in contrast to 'gold standard' DCs, which secrete little IL-12p70 and as a result induce mainly Th2 immunity, 'improved' cytokine cocktail DCs secreted higher levels IL-12p70 and also secreted similar concentration IL-10. To removal of PGE2 from the 'improved' DCs did increase the IL-12p70 production. In conclusion, we here present the 'improved' DCs, as an optimal maturation cocktail protocol, can induce high migratory potential, generate immunostimulatory DCs, produce higher levels IL-12p70 with superior capacity to induce Th1 immunity, when compared with the 'gold standard' DCs.
Collapse
Affiliation(s)
- Mei Zhu
- a Department of Laboratory Medicine ; Affiliated Provincial Hospital of Anhui Medical University ; Hefei , Anhui , China
| | | | | | | | | |
Collapse
|
11
|
Pitzler L, Auler M, Probst K, Frie C, Bergmeier V, Holzer T, Belluoccio D, van den Bergen J, Etich J, Ehlen H, Zhou Z, Bielke W, Pöschl E, Paulsson M, Brachvogel B. miR-126-3p Promotes Matrix-Dependent Perivascular Cell Attachment, Migration and Intercellular Interaction. Stem Cells 2016; 34:1297-309. [DOI: 10.1002/stem.2308] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/09/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Lena Pitzler
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Markus Auler
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Kristina Probst
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Christian Frie
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Vera Bergmeier
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Tatjana Holzer
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Daniele Belluoccio
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Cologne Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging - Associated Diseases (CECAD); University of Cologne; Cologne Germany
| | - Jocelyn van den Bergen
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Cologne Germany
- Murdoch Children's Research Institute; University of Melbourne; Parkville Victoria Australia
| | - Julia Etich
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Harald Ehlen
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Zhigang Zhou
- Department of Biochemistry and Molecular Biology; University of Melbourne; Parkville Victoria Australia
| | | | - Ernst Pöschl
- Department of Biochemistry and Molecular Biology; University of Melbourne; Parkville Victoria Australia
| | - Mats Paulsson
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
- Department of Paediatrics; University of Melbourne; Parkville Victoria Australia
- Norwich Medical School; University of East Anglia; Norwich Research Park Norwich United Kingdom
| | - Bent Brachvogel
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| |
Collapse
|
12
|
A20 regulates IL-1-induced tolerant production of CXC chemokines in human mesangial cells via inhibition of MAPK signaling. Sci Rep 2015; 5:18007. [PMID: 26648169 PMCID: PMC4673611 DOI: 10.1038/srep18007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023] Open
Abstract
Chemokines and chemokine receptors are involved in the resolution or progression of renal diseases. Locally secreted chemokines mediated leukocyte recruitment during the initiation and amplification phase of renal inflammation. However, the regulation of chemokine induction is not fully understood. In this study, we found that IL-1 induced a significant up-regulation of CXC chemokines CXCL1, 2, and 8 at both mRNA and protein levels in human mesangial cells. The induction of chemokines was tolerant, as the pre-treatment of HMC with IL-1 down-regulated the induction of chemokines induced by IL-1 re-stimulation. IL-1 up-regulated the ubiquintin-editing enzyme A20. A20 over-expression down-regulated IL-1-induced up-regulation of chemokines, and A20 down-regulation reversed chemokine inhibition induced by IL-1 pre-treatment, suggested that A20 played important roles in the tolerant production of chemokines. Unexpectedly, A20 over- expression inhibited the activation of ERK, JNK, and P38, but did not inhibit the activation of NF-κB. In addition, both IL-1 treatment and A20 over-expression induced the degradation of IRAK1, an important adaptor for IL-1R1 signaling, and A20 inhibition by RNA interference partly reversed the degradation of IRAK1. Taken together, IL-1-induced A20 negatively regulated chemokine production, suggesting that A20 may be an important target for the prevention and control of kidney inflammation.
Collapse
|
13
|
Chen Y, Zhou S, Hua K, Xiao H, Li Z, Liu M, Luo R, Bi D, Zhou R, Jin H. Haemophilus parasuis infection activates chemokine RANTES in PK-15 cells. Mol Immunol 2015. [PMID: 26198698 DOI: 10.1016/j.molimm.2015.06.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
RANTES is a member of the CC chemokine involved in inflammation and immune response during pathogen infection. In our previous study, Haemophilus parasuis (H. parasuis), which is responsible for the great economic losses in the pig industry worldwide, has been shown to enhance RANTES expression in PK-15 cells. However, the mechanisms behind this biological phenomenon have remained unclear. In this study, we showed that H. parasuis infection significantly upregulated RANTES gene transcription in a time- and dose-dependent manner. Promoter analysis by site-directed mutagenesis indicated that the nuclear factor NF-κB binding site was the most important cis-regulatory element controlling H. parasuis-induced RANTES transcription. Inhibition of NF-κB and JNK activity also significantly reduced H. parasuis-induced RANTES production. In addition, TLRs signaling pathway was found to be involved in H. parasuis induced-RANTES expression. These results represent an important molecular mechanism whereby H. parasuis induced RANTES in the inflammatory response.
Collapse
Affiliation(s)
- Yushan Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shanshan Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Kexin Hua
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongde Xiao
- Hubei Center for Animal Disease Control and Prevention, Wuhan 430070, China
| | - Zili Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Mei Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Dingren Bi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
14
|
Wu HM, Fang L, Shen QY, Liu RY. SP600125 promotes resolution of allergic airway inflammation via TLR9 in an OVA-induced murine acute asthma model. Mol Immunol 2015; 67:311-6. [PMID: 26139014 DOI: 10.1016/j.molimm.2015.06.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND c-Jun N-terminal kinase (JNK) relays extracellular stimuli through phosphorylation cascades that lead to various cell responses. In the present study, we aimed to investigate the effect of the JNK inhibitor SP600125 on the resolution of airway inflammation, and the underlying mechanism using a murine acute asthma model. METHODS Female C57BL/6 mice were sensitized with saline or ovalbumin (OVA) on day 0, and challenged with OVA on day 14-20. Meanwhile, some of the mice were treated with SP600125 (30 mg/kg) intraperitoneally 2 h before each challenge. The airway inflammation was evaluated by counting the numbers of various types of inflammatory cells in bronchoalveolar lavage fluid (BALF), histopathology, cytokines production and mucus secretion in individual mouse. In addition, we analyzed the protein levels of phosphorylated JNK and TLR9 in the lung tissues. RESULTS SP600125 markedly reduced the invasion of inflammatory cells into the peribronchial regions, and decreased the numbers of eosinophils, monocytes, neutrophils and lymphocytes in BALF. SP600125 also reduced the level of plasma OVA-specific IgE, lowered the production of pro-inflammatory cytokines in BALF and alleviated mucus secretion. Meanwhile, SP600125 inhibited OVA-induced, increased expression of p-JNK and TLR9 in the lung tissues. CONCLUSIONS Collectively, our data demonstrated that SP600125 promoted resolution of allergic airway inflammation via TLR9 in an OVA-induced murine acute asthma model. The JNK-TLR9 pathway may be a new therapeutic target in the treatment for the allergic asthma.
Collapse
Affiliation(s)
- Hui-Mei Wu
- Anhui Geriatric Institute, Department of Pulmonary, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Lei Fang
- Anhui Geriatric Institute, Department of Pulmonary, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Qi-Ying Shen
- Anhui Geriatric Institute, Department of Pulmonary, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Rong-Yu Liu
- Anhui Geriatric Institute, Department of Pulmonary, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China.
| |
Collapse
|
15
|
Jiang M, Broering R, Trippler M, Poggenpohl L, Fiedler M, Gerken G, Lu M, Schlaak JF. Toll-like receptor-mediated immune responses are attenuated in the presence of high levels of hepatitis B virus surface antigen. J Viral Hepat 2014; 21:860-72. [PMID: 24498958 DOI: 10.1111/jvh.12216] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/08/2013] [Indexed: 12/13/2022]
Abstract
It has been recently shown that Toll-like receptor (TLR) signalling in murine nonparenchymal liver cells (NPCs) is suppressed in the presence of Hepatitis B virus surface antigen (HBsAg). It is not clear, however, whether this is also relevant for the adaptive immune responses and how this effect is mediated. Peripheral blood mononuclear cells (PBMCs) from Hepatitis B virus (HBV) patients and controls were stimulated by TLR ligands in the absence or presence of autologous serum. Interestingly, TLR-mediated cytokine expression (Interleukin-6 and -10) as well as TLR3-induced interferon (IFN) expression in PBMCs of HBV patients was significantly higher than in the healthy volunteers, showing a negative correlation with the levels of HBsAg. In addition, TLR3-mediated IFN-γ production was inhibited in the presence of HBV-containing serum. To mechanistically analyse this observation, murine Kupffer cells (KCs) and sinusoidal endothelial cells (LSECs) were stimulated with TLR3 ligands in the presence or absence of HBsAg. Mixed lymphocyte reactions were performed to study T-cell activation induced by TLR-stimulated NPCs. Gene expression of cytokines and TLR3 was analysed by quantitative rt-PCR, and activation of transcription factors was assessed by Western blot or reporter gene assays. TLR-induced expression of interferon γ, interferon sensitive genes and proinflammatory cytokines in murine KCs and LSECs was efficiently suppressed in the presence of HBsAg, whereas the expression of anti-inflammatory cytokines was enhanced. Activation of NFκB, IRF-3 and MAPKs in these liver cells was potently suppressed by HBsAg. T-cell activation mediated through TLR3-stimulated KCs or LSECs was suppressed by HBsAg which could be reverted by anti-IL-10 antibodies. These findings may, at least in part, explain how HBV evades innate and adaptive immune responses to maintain a persistent infection.
Collapse
Affiliation(s)
- M Jiang
- Faculty of Medicine, Department of Gastroenterology and Hepatology, University Duisburg-Essen, Essen, Germany; Faculty of Medicine, Institute of Virology, University Duisburg-Essen, Essen, Germany; Department of Pathogenic Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Li L, Cheng FW, Wang F, Jia B, Luo X, Zhang SQ. The activation of TLR7 regulates the expression of VEGF, TIMP1, MMP2, IL-6, and IL-15 in Hela cells. Mol Cell Biochem 2013; 389:43-9. [PMID: 24347177 DOI: 10.1007/s11010-013-1925-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/06/2013] [Indexed: 12/31/2022]
Abstract
Toll-like receptors (TLRs) play important roles in activation of immunoreaction and tumor development. Toll-like receptor 7 (TLR7), one of the TLRs binding with single-stranded RNA, activates intracellular pathways and stimulates the release of proinflammatory cytokines, chemokines. In this study, we investigated the impact of the TLR7-signaling pathway on the expression of vascular endothelial growth factor (VEGF), matrix metalloproteinase 2 (MMP2), tissue inhibitor of metalloproteinase 1 (TIMP1), interleukin 6 (IL-6), and interleukin 15 (IL-15), which have been testified to refer to the immunomodulating and tumor progression. We confirmed that the TLR7 was expressed by Hela cells, despite the abundance was weak. Gardiquimod, one of the TLR7 ligands, can promote these five genes expression in varying degrees. After stimulating with gardiquimod, the expression of the IL-15V1, 3 increased about 4.5 times on RNA level, the other expression was only up-regulated about 2 times. We also discovered that gardiquimod could activate the MAPK/ERK- and PI3K/AKT-signaling pathways, and the specific inhibitors studies indicate that, the effect of gardiquimod on these genes expression is mainly or partially dependent on the activation of these two signaling pathways. To sum up, the activation of TLR7 signaling pathway may modulate some genes expression in Hela cells and may contribute to the pathogenesis of the cervical cancer.
Collapse
Affiliation(s)
- Lei Li
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui, China
| | | | | | | | | | | |
Collapse
|
17
|
Jia B, Luo X, Cheng FW, Li L, Hu DJ, Wang F, Zhang SQ. Gardiquimod inhibits the expression of calcium-induced differentiation markers in HaCaT cells. Mol Biol Rep 2013; 40:6363-9. [PMID: 24057248 DOI: 10.1007/s11033-013-2750-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 09/14/2013] [Indexed: 10/26/2022]
Abstract
Toll-like receptor 7 (TLR7) is an important member in pattern recognition receptors families. TLR7 signal pathway is involved in the physiological process in many type cells, but the impact of TRL7 on differentiation in the human keratinocytes is still unknown. In this study, we investigated the expression of TLR7 in keratinocytes, and the effect of TLR7 agonist gardiquimod on the expression of calcium (Ca(2+))-induced keratinocytes differentiation markers in HaCaT cells. Immunohistochemistry and western-blotting analysis showed that TLR7 is expressed in basal keratinocytes of normal skin and in the human keratinocyte cell line HaCaT, but not expressed in the keratinocytes of psoriasis lesions. Pretreatment with gardiquimod could down-regulate Ca(2+)-induced differentiation marker expression and activate Raf-MEK-ERK and PI3K-AKT signal pathways in HaCaT cells. However, specific inhibitors studies showed that the down-regulation of the differentiation markers expression by gardiquimod was not dependent on the activation of these two pathways. TLR7 may play an important role in the pathogenesis of psoriasis through regulating the differentiation of the keratinocytes, and will give a new insight into the psoriasis.
Collapse
Affiliation(s)
- Bo Jia
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Brieger A, Rink L, Haase H. Differential regulation of TLR-dependent MyD88 and TRIF signaling pathways by free zinc ions. THE JOURNAL OF IMMUNOLOGY 2013; 191:1808-17. [PMID: 23863901 DOI: 10.4049/jimmunol.1301261] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Zinc signals are utilized by several immune cell receptors. One is TLR4, which causes an increase of free zinc ions (Zn(2+)) that is required for the MyD88-dependent expression of inflammatory cytokines. This study investigates the role of Zn(2+) on Toll/IL-1R domain-containing adapter inducing IFN-β (TRIF)-dependent signals, the other major intracellular pathway activated by TLR4. Chelation of Zn(2+) with the membrane-permeable chelator N,N,N',N'-Tetrakis(2-pyridylmethyl)ethylenediamine augmented TLR4-mediated production of IFN-β and subsequent synthesis of inducible NO synthase and production of NO. The effect is based on Zn(2+) acting as a negative regulator of the TRIF pathway via reducing IFN regulatory factor 3 activation. This was also observed with TLR3, the only TLR that signals exclusively via TRIF, but not MyD88, and does not trigger a zinc signal. In contrast, IFN-γ-induced NO production was unaffected by N,N,N',N'-Tetrakis(2-pyridylmethyl)ethylenediamine. Taken together, Zn(2+) is specifically involved in TLR signaling, where it differentially regulates MyD88 and TRIF signaling via a zinc signal or via basal Zn(2+) levels, respectively.
Collapse
Affiliation(s)
- Anne Brieger
- Institute of Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | | | | |
Collapse
|
19
|
Group B Streptococcus and Streptococcus suis capsular polysaccharides induce chemokine production by dendritic cells via Toll-like receptor 2- and MyD88-dependent and -independent pathways. Infect Immun 2013; 81:3106-18. [PMID: 23774593 DOI: 10.1128/iai.00113-13] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Streptococcus agalactiae (also known as group B Streptococcus [GBS]) and Streptococcus suis are encapsulated streptococci causing severe septicemia and meningitis. Bacterial capsular polysaccharides (CPSs) are poorly immunogenic, but anti-CPS antibodies are essential to the host defense against encapsulated bacteria. The mechanisms underlying anti-CPS antibody responses are not fully elucidated, but the biochemistry of CPSs, particularly the presence of sialic acid, may have an immunosuppressive effect. We investigated the ability of highly purified S. suis and GBS native (sialylated) CPSs to activate dendritic cells (DCs), which are crucial actors in the initiation of humoral immunity. The influence of CPS biochemistry was studied using CPSs extracted from different serotypes within these two streptococcal species, as well as desialylated CPSs. No interleukin-1β (IL-1β), IL-6, IL-12p70, tumor necrosis factor alpha (TNF-α), or IL-10 production was observed in S. suis or GBS CPS-stimulated DCs. Moreover, these CPSs exerted immunosuppressive effects on DC activation, as a diminution of gamma interferon (IFN-γ)-induced B cell-activating factor of the tumor necrosis factor family (BAFF) expression was observed in CPS-pretreated cells. However, S. suis and GBS CPSs induced significant production of CCL3, via partially Toll-like receptor 2 (TLR2)- and myeloid differentiation factor 88 (MyD88)-dependent pathways, and CCL2, via TLR-independent mechanisms. No major influence of CPS biochemistry was observed on the capacity to induce chemokine production by DCs, indicating that DCs respond to these CPSs in a patterned way rather than a structure-dedicated manner.
Collapse
|
20
|
Klettner A, Koinzer S, Meyer T, Roider J. Toll-like receptor 3 activation in retinal pigment epithelium cells - Mitogen-activated protein kinase pathways of cell death and vascular endothelial growth factor secretion. Acta Ophthalmol 2013; 91:e211-8. [PMID: 23387336 DOI: 10.1111/aos.12031] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Toll-like receptor 3 (TLR3) is a receptor of the innate immune system, recognizing double-stranded RNA. TLR3 can lead to cytokine release or apoptosis and has recently been associated with the development of geographical atrophy via cytotoxic effects on the retinal pigment epithelium (RPE). The current study was conducted to elucidate the underlying pathways of TLR3 effects in the RPE. METHODS TLR3 activation via polyinosinic acid/polycytidylic acid (Poly I:C) was investigated in primary porcine RPE cells, focussing on cell death and vascular endothelial growth factor (VEGF) secretion. Primary cells were stimulated with different concentrations of Poly I:C. Cell death was investigated in trypan blue exclusion assay and cell death detection ELISA. VEGF and IFN-ß secretion were also detected in ELISA. As Mitogen-activated protein kinases (MAPK) play an important part in TLR3-mediated signal transduction, we investigated the influence of JNK, ERK1/2 and p38 on cell death and VEGF secretion, using commercially available inhibitors. RESULTS Activation of TLR3 by Poly I:C induced concentration-dependent cell death, partly mediated by JNK. ERK1/2 was activated and exerted some protection. Furthermore, higher concentrations of Poly I:C increased VEGF secretion after 4 and 24 hr, which was independent of MAPK. CONCLUSION The induction of cell death in RPE cells by TLR3 activation confirms possible involvement of TLR3 activation in GA. As cell death is partly mediated by JNK, more studies should be conducted investigating the role of JNK in RPE cell death to evaluate whether its inhibition might be a new therapeutic opportunity for the treatment of geographical atrophy. Additionally, effects on VEGF secretion can be found.
Collapse
Affiliation(s)
- Alexa Klettner
- Department of Ophthalmology, University of Kiel, Kiel, Germany.
| | | | | | | |
Collapse
|
21
|
The innate immune response to hepatitis B virus infection: Implications for pathogenesis and therapy. Antiviral Res 2012; 96:405-13. [DOI: 10.1016/j.antiviral.2012.10.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 10/02/2012] [Accepted: 10/05/2012] [Indexed: 02/07/2023]
|
22
|
Guo F, Mead J, Aliya N, Wang L, Cuconati A, Wei L, Li K, Block TM, Guo JT, Chang J. RO 90-7501 enhances TLR3 and RLR agonist induced antiviral response. PLoS One 2012; 7:e42583. [PMID: 23056170 PMCID: PMC3463586 DOI: 10.1371/journal.pone.0042583] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/09/2012] [Indexed: 12/20/2022] Open
Abstract
Recognition of virus infection by innate pattern recognition receptors (PRRs), including membrane-associated toll-like receptors (TLR) and cytoplasmic RIG-I-like receptors (RLR), activates cascades of signal transduction pathways leading to production of type I interferons (IFN) and proinflammatory cytokines that orchestrate the elimination of the viruses. Although it has been demonstrated that PRR-mediated innate immunity plays an essential role in defending virus from infection, it also occasionally results in overwhelming production of proinflammatory cytokines that cause severe inflammation, blood vessel leakage and tissue damage. In our efforts to identify small molecules that selectively enhance PRR-mediated antiviral, but not the detrimental inflammatory response, we discovered a compound, RO 90-7501 ('2'-(4-Aminophenyl)-[2,5'-bi-1H-benzimidazol]-5-amine), that significantly promoted both TLR3 and RLR ligand-induced IFN-β gene expression and antiviral response, most likely via selective activation of p38 mitogen-activated protein kinase (MAPK) pathway. Our results thus imply that pharmacological modulation of PRR signal transduction pathways in favor of the induction of a beneficial antiviral response can be a novel therapeutic strategy.
Collapse
Affiliation(s)
- Fang Guo
- Department of Microbiology and Immunology, Drexel University College of Medicine, Doylestown, Pennsylvania, United States of America
| | - Jennifer Mead
- Department of Microbiology and Immunology, Drexel University College of Medicine, Doylestown, Pennsylvania, United States of America
| | - Nishat Aliya
- Department of Microbiology and Immunology, Drexel University College of Medicine, Doylestown, Pennsylvania, United States of America
| | - Lijuan Wang
- Department of Microbiology and Immunology, Drexel University College of Medicine, Doylestown, Pennsylvania, United States of America
| | - Andrea Cuconati
- Institute for Hepatitis Virus Research, Hepatitis B Foundation, Doylestown, Pennsylvania, United States of America
| | - Lai Wei
- Institute of Hepatology, Peking University, Beijing, China
| | - Kui Li
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Timothy M. Block
- Department of Microbiology and Immunology, Drexel University College of Medicine, Doylestown, Pennsylvania, United States of America
- Institute for Hepatitis Virus Research, Hepatitis B Foundation, Doylestown, Pennsylvania, United States of America
| | - Ju-Tao Guo
- Department of Microbiology and Immunology, Drexel University College of Medicine, Doylestown, Pennsylvania, United States of America
| | - Jinhong Chang
- Department of Microbiology and Immunology, Drexel University College of Medicine, Doylestown, Pennsylvania, United States of America
| |
Collapse
|
23
|
Ortiz-Stern A, Deng X, Smoktunowicz N, Mercer PF, Chambers RC. PAR-1-dependent and PAR-independent pro-inflammatory signaling in human lung fibroblasts exposed to thrombin. J Cell Physiol 2012; 227:3575-84. [PMID: 22278285 DOI: 10.1002/jcp.24061] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Proteinase-activated receptors (PARs) are crucial in orchestrating cellular responses to coagulation proteinases, such as thrombin and FXa. Four PARs have been characterized and have been shown to be differentially expressed in mice and humans and between tissues. We have previously shown that in murine lung fibroblasts, PAR-1 is solely responsible for all cellular responses to thrombin and FXa. In contrast, we report here that in primary human lung fibroblasts (pHLFs), known PARs fail to account for all of the cellular responses to thrombin, in particular in the presence of high, but physiologically achievable concentrations of thrombin. We report that pHLFs secrete CCL2 in a PAR-1-dependent manner at low thrombin concentration (∼0.3 nM). At or above 10 nM thrombin, pharmacological antagonism (RWJ-58259) fails to block thrombin-induced CCL2 release; whereas PAR-1 cleavage-blocking monoclonal antibodies (ATAP2 and WEDE15) only partially inhibit thrombin-induced CCL2 secretion. In addition, activation of PAR-3, PAR-4, and transactivation of either PAR-2 or EGFR were ruled out as being responsible for thrombin-mediated CCL2 secretion at high yet standard concentrations of the proteinase. We further provide evidence that PAR-1-dependent and PAR-independent signaling involves the rapid phosphorylation of ERK, which in turn is absolutely required for thrombin-induced CCL2 secretion at both low and standard concentration of the proteinase. Our findings suggest the existence of a PAR-independent signaling mechanism in human lung fibroblasts and have important implications for the design of therapeutic strategies aimed at blocking pro-inflammatory signaling responses associated with excessive thrombin generation.
Collapse
|
24
|
Fairman P, Angel JB. The effect of human immunodeficiency virus-1 on monocyte-derived dendritic cell maturation and function. Clin Exp Immunol 2012; 170:101-13. [PMID: 22943206 PMCID: PMC3444722 DOI: 10.1111/j.1365-2249.2012.04628.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2012] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DC) are mediators of the adaptive immune response responsible for antigen presentation to naive T cells in secondary lymph organs. Human immunodeficiency virus (HIV-1) has been reported to inhibit the maturation of DC, but a clear link between maturation and function has not been elucidated. To understand further the effects of HIV-1 on DC maturation and function, we expanded upon previous investigations and assessed the effects of HIV-1 infection on the expression of surface molecules, carbohydrate endocytosis, antigen presentation and lipopolysaccharide (LPS) responsiveness over the course of maturation. In vitro infection with HIV-1 resulted in an increase in the expression of DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) as well as decreases in maturation-induced CCR7 and major histocompatibility complex (MHC)-II expression. Retention of endocytosis that normally occurs with DC maturation as well as inhibition of antigen presentation to CD8(+) T cells was also observed. Mitogen-activated protein kinase (MAPK) responsiveness to LPS as measured by phosphorylation of p38, c-Jun N-terminal kinase (JNK) and extracellular-regulated kinase (ERK)1/2 was not affected by HIV-1 infection. In summary, in-vitro HIV-1 impairs DC maturation, as defined by cell surface protein expression, with selective alterations in mature DC function. Understanding the mechanisms of DC dysfunction in HIV infection will provide further insight into HIV immune pathogenesis.
Collapse
Affiliation(s)
- P Fairman
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | |
Collapse
|
25
|
Forrest CM, Khalil OS, Pisar M, Smith RA, Darlington LG, Stone TW. Prenatal activation of Toll-like receptors-3 by administration of the viral mimetic poly(I:C) changes synaptic proteins, N-methyl-D-aspartate receptors and neurogenesis markers in offspring. Mol Brain 2012; 5:22. [PMID: 22681877 PMCID: PMC3496691 DOI: 10.1186/1756-6606-5-22] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/25/2012] [Indexed: 12/30/2022] Open
Abstract
Background There is mounting evidence for a neurodevelopmental basis for disorders such as autism and schizophrenia, in which prenatal or early postnatal events may influence brain development and predispose the young to develop these and related disorders. We have now investigated the effect of a prenatal immune challenge on brain development in the offspring. Pregnant rats were treated with the double-stranded RNA polyinosinic:polycytidylic acid (poly(I:C); 10 mg/kg) which mimics immune activation occurring after activation of Toll-like receptors-3 (TLR3) by viral infection. Injections were made in late gestation (embryonic days E14, E16 and E18), after which parturition proceeded naturally and the young were allowed to develop up to the time of weaning at postnatal day 21 (P21). The brains of these animals were then removed to assess the expression of 13 different neurodevelopmental molecules by immunoblotting. Results Measurement of cytokine levels in the maternal blood 5 hours after an injection of poly(I:C) showed significantly increased levels of monocyte chemoattractant protein-1 (MCP-1), confirming immune activation. In the P21 offspring, significant changes were detected in the expression of GluN1 subunits of NMDA receptors, with no difference in GluN2A or GluN2B subunits or the postsynaptic density protein PSD-95 and no change in the levels of the related small GTPases RhoA or RhoB, or the NMDA receptor modulator EphA4. Among presynaptic molecules, a significant increase in Vesicle Associated Membrane Protein-1 (VAMP-1; synaptobrevin) was seen, with no change in synaptophysin or synaptotagmin. Proliferating Cell Nuclear Antigen (PCNA), as well as the neurogenesis marker doublecortin were unchanged, although Sox-2 levels were increased, suggesting possible changes in the rate of new cell differentiation. Conclusions The results reveal the induction by prenatal poly(I:C) of selective molecular changes in the brains of P21 offspring, affecting primarily molecules associated with neuronal development and synaptic transmission. These changes may contribute to the behavioural abnormalities that have been reported in adult animals after exposure to poly(I:C) and which resemble symptoms seen in schizophrenia and related disorders.
Collapse
Affiliation(s)
- Caroline M Forrest
- Institute for Neuroscience and Psychology, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | | | | | | | | | | |
Collapse
|
26
|
Johnson DS, Chen YH. Ras family of small GTPases in immunity and inflammation. Curr Opin Pharmacol 2012; 12:458-63. [PMID: 22401931 DOI: 10.1016/j.coph.2012.02.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 02/02/2012] [Accepted: 02/03/2012] [Indexed: 11/29/2022]
Abstract
The Ras superfamily of small GTPases is a group of more than 150 small G proteins, all of which share some degree of homology to the founding member Ras. These small GTPases function as molecular switches within cells, impacting nearly all cellular processes. The Ras superfamily can be further divided into several smaller subfamilies, with those proteins that most closely resemble Ras belonging to the Ras subfamily. While heavily studied within the field of cancer biology, the Ras family of proteins also plays cardinal roles in immunity and inflammation. Here we review the roles of these molecular switches in regulating immune cell homeostasis and functions.
Collapse
Affiliation(s)
- Derek S Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
27
|
Ning R, Zhang X, Guo X, Li Q. Staphylococcus aureus regulates secretion of interleukin-6 and monocyte chemoattractant protein-1 through activation of nuclear factor kappaB signaling pathway in human osteoblasts. Braz J Infect Dis 2011; 15:189-94. [PMID: 21670915 DOI: 10.1016/s1413-8670(11)70173-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 09/16/2010] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE Activation of nuclear factor kappaB by diverse bacteria regulates the secretion of chemokines and cytokines. Staphylococcus aureus (S. aureus)-infected osteoblasts can significantly increase the secretion of interleukin-6 and monocyte chemoattractant protein-1. The aim of this study was to investigate whether S. aureus can activate nuclear factor kappaB in human osteoblasts, and whether the activation of nuclear factor kappaB by S. aureus regulates the secretion of interleukin-6 and monocyte chemoattractant protein-1. METHODS Immunoblot and electrophoretic mobility shift assay were used to detect the degradation of IκBa and activation of nuclear factor kappaB in human osteoblasts in response to S. aureus, respectively. Enzyme-linked immunosorbent assay was used to measure the secretion of interleukin-6 and monocyte chemoattractant protein-1 in the supernatants. Lastly, carbobenzoxyl-l-leucinyl-l-leucinyl-l-leucinal, an inhibitor of the nuclear factor kappaB, was used to determine if activation of nuclear factor kappaB by S. aureus in human osteoblasts regulates the secretions of interleukin-6 and monocyte chemoattractant protein-1. RESULTS Our results for the first time demonstrated that S. aureus can induce the degradation of IκBa and activation of nuclear factor kappaB in human osteoblasts in a time and dose-dependent manner. In addition, inhibition of nuclear factor kappaB by carbobenzoxyl-l-leucinyl-l-leucinyl-l-leucinal suppressed the secretion of interleukin-6 and monocyte chemoattractant protein-1 in the supernatants of S. aureus-infected human osteoblasts in a dose-dependent manner. CONCLUSION These findings suggest that S. aureus can activate nuclear factor kappaB in human osteoblasts, and subsequently regulate the secretion of interleukin-6 and monocyte chemoattractant protein-1. The nuclear factor kappaB transcription factor regulates a number of genes involved in a wide variety of biological processes. Further study of the effects of nuclear factor kappaB activation on S. aureus-infected human osteoblast may provide us new insights into discovery of the immune mechanisms in osteomyelitis.
Collapse
Affiliation(s)
- Rende Ning
- Department of Orthopaedics, The Sixth People's Hospital Affiliated, Shanghai Jiao Tong University, China
| | | | | | | |
Collapse
|
28
|
Di Caro V, D'Anneo A, Phillips B, Engman C, Harnaha J, Trucco M, Giannoukakis N. Phosphatidylinositol-3-kinase activity during in vitro dendritic cell generation determines suppressive or stimulatory capacity. Immunol Res 2011; 50:130-52. [PMID: 21476100 DOI: 10.1007/s12026-011-8206-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Modulating PI3K at different stages of dendritic cells (DC) generation could be a novel means to balance the generation of immunosuppressive versus immunostimulatory DC. We show that PI3K inhibition during mouse DC generation in vitro results in cells that are potently immunosuppressive and characteristic of CD8alpha- CD11c+ CD11b+ DC. These DC exhibited low surface class I and class II MHC, CD40, and CD86 and did not produce TNF-alpha. In allogeneic MLR, these DC were suppressive. Although in these mixed cultures, there was no increase in the frequency of CD4+ CD25+ Foxp3+ cells, the Foxp3 content on a per cell basis was significantly increased. Sustained TLR9 signaling in the presence of PI3K inhibition during DC generation overrode the cells' suppressive phenotype.
Collapse
Affiliation(s)
- Valentina Di Caro
- Department of Pediatrics, Division of Immunogenetics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Kim ES, Chang YS, Choi SJ, Kim JK, Yoo HS, Ahn SY, Sung DK, Kim SY, Park YR, Park WS. Intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells attenuates Escherichia coli-induced acute lung injury in mice. Respir Res 2011; 12:108. [PMID: 21843339 PMCID: PMC3166924 DOI: 10.1186/1465-9921-12-108] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 08/15/2011] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Human umbilical cord blood (UCB)-derived mesenchymal stem cells (MSCs) attenuate hyperoxic neonatal lung injury primarily through anti-inflammatory effects. We hypothesized that intratracheal transplantation of human UCB-derived MSCs could attenuate Escherichia coli (E. coli)-induced acute lung injury (ALI) in mice by suppressing the inflammatory response. METHODS Eight-week-old male ICR mice were randomized to control or ALI groups. ALI was induced by intratracheal E. coli instillation. Three-hours after E. coli instillation, MSCs, fibroblasts or phosphate-buffered saline were intratracheally administered randomly and survival was analyzed for 7 days post-injury. Lung histology including injury scores, myeloperoxidase (MPO) activity, and protein levels of interleukin (IL)-1α, IL-1β, IL-6, tumor necrosis factor (TNF)-α, and macrophage inflammatory protein (MIP)-2 as well as the wet-dry lung ratio and bacterial counts from blood and bronchoalveolar lavage (BAL) were evaluated at 1, 3, and 7 days post-injury. Levels of inflammatory cytokines in the lung were also profiled using protein macroarrays at day 3 post-injury which showed peak inflammation. RESULTS MSC transplantation increased survival and attenuated lung injuries in ALI mice, as evidenced by decreased injury scores on day 3 post-injury and reduced lung inflammation including increased MPO activity and protein levels of IL-1α, IL-1β, IL-6, TNF-α, and MIP-2 on day 3 and 7 post-injury. Inflammatory cytokine profiles in the lungs at day 3 post-injury were attenuated by MSC transplantation. MSCs also reduced the elevated lung water content at day 3 post-injury and bacterial counts in blood and BAL on day 7 post-injury. CONCLUSIONS Intratracheal transplantation of UCB-derived MSCs attenuates E. coli-induced ALI primarily by down-modulating the inflammatory process and enhancing bacterial clearance.
Collapse
Affiliation(s)
- Eun Sun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Specific immunotherapy and turning off the T cell: how does it work? Ann Allergy Asthma Immunol 2011; 107:381-92. [PMID: 22018608 DOI: 10.1016/j.anai.2011.05.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 03/08/2011] [Accepted: 05/17/2011] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To examine T-regulatory (Treg) cell functions in allergic immune responses and their roles during allergen specific immunotherapy based on recent developments and current understanding of immune regulation. DATA SOURCES PubMed search of English-language articles regarding Treg cells and allergen specific immunotherapy. STUDY SELECTION Articles on the subject matter were selected and reviewed. RESULTS Allergen specific immunotherapy is the ultimate treatment modality targeting the immunopathogenic mechanisms of allergic disorders. A diminished allergen-specific T-cell proliferation and suppressed secretion of T(H)1- and T(H)2-type cytokines are the characteristic hallmarks. In addition, Treg cells inhibit the development of allergen-specific T(H)2 and T(H)1 cell responses and therefore exert key roles in healthy immune response to allergens. Treg cells potently suppress IgE production and directly or indirectly control the activity of effector cells of allergic inflammation, such as eosinophils, basophils, and mast cells. CONCLUSION As advancements in the field of allergen specific immunotherapy ensue, they may provide novel progression of more rational and safer approaches for the prevention and treatment of allergic disorders. Currently, the Treg cell field is an open research area to increase our understanding in mechanisms of peripheral tolerance to allergens.
Collapse
Affiliation(s)
- Cevdet Ozdemir
- Division of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
| | | | | | | |
Collapse
|