1
|
Astragalus Saponins, Astragaloside VII and Newly Synthesized Derivatives, Induce Dendritic Cell Maturation and T Cell Activation. Vaccines (Basel) 2023; 11:vaccines11030495. [PMID: 36992079 DOI: 10.3390/vaccines11030495] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Astragaloside VII (AST VII), a triterpenic saponin isolated from Astragalus species, shows promise as a vaccine adjuvant, as it supported a balanced Th1/Th2 immune response in previous in vivo studies. However, the underlying mechanisms of its adjuvant activity have not been defined. Here, we investigated the impact of AST VII and its newly synthesized semi-synthetic analogs on human whole blood cells, as well as on mouse bone marrow-derived dendritic cells (BMDCs). Cells were stimulated with AST VII and its derivatives in the presence or absence of LPS or PMA/ionomycin and the secretion of cytokines and the expression of activation markers were analyzed using ELISA and flow cytometry, respectively. AST VII and its analogs increased the production of IL-1β in PMA/ionomycin-stimulated human whole blood cells. In LPS-treated mouse BMDCs, AST VII increased the production of IL-1β and IL-12, and the expression of MHC II, CD86, and CD80. In mixed leukocyte reaction, AST VII and derivatives increased the expression of the activation marker CD44 on mouse CD4+ and CD8+ T cells. In conclusion, AST VII and its derivatives strengthen pro-inflammatory responses and support dendritic cell maturation and T cell activation in vitro. Our results provide insights into the mechanisms of the adjuvant activities of AST VII and its analogs, which will be instrumental to improve their utility as a vaccine adjuvant.
Collapse
|
2
|
Wang M, Zhong J, Xiang Y. LncRNA‐GAS5 related to the processes of recurrent pregnancy loss by regulating Th1/Th2 balance. Kaohsiung J Med Sci 2021; 37:479-486. [PMID: 33511769 DOI: 10.1002/kjm2.12360] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
- Miao‐Miao Wang
- Department of Reproductive Center Huai'an Maternal and Child Health Care Hospital, Xuzhou Medical University Huai'an Jiangsu China
| | - Ji‐Xiang Zhong
- Department of gynaecology Huai'an Maternal and Child Health Care Hospital, Xuzhou Medical University Huai'an Jiangsu China
| | - Yuan‐Yuan Xiang
- Department of gynaecology Huai'an Maternal and Child Health Care Hospital, Xuzhou Medical University Huai'an Jiangsu China
| |
Collapse
|
3
|
Moerman-Herzog AM, Acheampong DA, Brooks AG, Blair SM, Hsu PC, Wong HK. Transcriptome analysis of Sézary syndrome and lymphocytic-variant hypereosinophilic syndrome T cells reveals common and divergent genes. Oncotarget 2019; 10:5052-5069. [PMID: 31489115 PMCID: PMC6707948 DOI: 10.18632/oncotarget.27120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/15/2019] [Indexed: 11/25/2022] Open
Abstract
Sézary syndrome (SS) is an aggressive cutaneous T cell lymphoma with pruritic skin inflammation and immune dysfunction, driven by neoplastic, clonal memory T cells in both peripheral blood and skin. To gain insight into abnormal gene expression promoting T cell dysfunction, lymphoproliferation and transformation in SS, we first compared functional transcriptomic profiles of both resting and activated CD4+CD45RO+ T cells from SS patients and normal donors to identified differential expressed genes. Next, a meta-analysis was performed to compare our SS data to public microarray data from a novel benign disease control, lymphocytic-variant hypereosinophilic syndrome (L-HES). L-HES is a rare, clonal lymphoproliferation of abnormal memory T cells that produces similar clinical symptoms as SS, including severe pruritus and eosinophilia. Comparison revealed gene sets specific for either SS (370 genes) or L-HES (519 genes), and a subset of 163 genes that were dysregulated in both SS and L-HES T cells compared to normal donor T cells. Genes confirmed by RT-qPCR included elevated expression of PLS3, TWIST1 and TOX only in SS, while IL17RB mRNA was increased only in L-HES. CDCA7 was increased in both diseases. In an L-HES patient who progressed to peripheral T cell lymphoma, the malignant transformation identified increases in the expression of CDCA7, TIGIT, and TOX, which are highly expressed in SS, suggesting that these genes contribute to neoplastic transformation. In summary, we have identified gene expression biomarkers that implicate a common transformative mechanism and others that are unique to differentiate SS from L-HES.
Collapse
Affiliation(s)
- Andrea M Moerman-Herzog
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Daniel A Acheampong
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Joint Graduate Program in Bioinformatics, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amanda G Brooks
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Suzan M Blair
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ping-Ching Hsu
- Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Henry K Wong
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
4
|
Lin CY, Zu CH, Yang CC, Tsai PJ, Shyu JF, Chen CP, Weng ZC, Chen TH, Wang HS. IL-1β-Induced Mesenchymal Stem Cell Migration Involves MLCK Activation via PKC Signaling. Cell Transplant 2014; 24:2011-28. [PMID: 25333338 DOI: 10.3727/096368914x685258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Mesenchymal stem cells (MSCs) migrate via the bloodstream to sites of injury, possibly attracted by inflammatory cytokines. Although many cytokines can induce stem cell migration, the underlying mechanism is not fully understood. We found that tail vein-injected MSCs migrate to the pancreas in nonobese diabetic (NOD) mice. An ELISA assay revealed that hyperglycemic NOD mice have higher pancreatic levels of interleukin-1β (IL-1β) than normal NOD mice and that IL-1β stimulates MSC migration in a Transwell assay and electric cell-substrate impedance sensing system. Microarray analysis showed that myosin light chain kinase (MLCK) is involved in IL-1β-induced MSC migration, while Western blots showed that IL-1β stimulates MLCK expression and activation and that MLCK-siRNA transfection reduces MSC migration. Kinase inhibitors, chromatin immunoprecipitation, and a knockdown study revealed that IL-1β-induced MLCK expression is regulated by the PKCδ/NF-κB signaling pathway, and a kinase inhibitor study revealed that IL-1β-induced MLCK activation occurs via the PKCα/MEK/ERK signaling pathway. These results show that IL-1β released from the pancreas of hyperglycemic NOD mice induces MSC migration and that this is dependent on MLCK expression via the PKCδ/NF-κB pathway and on MLCK activation via the PKCα/MEK/ERK signaling cascade. This study increases our understanding of the mechanisms by which MSCs home to injury sites.
Collapse
Affiliation(s)
- Cheng-Yu Lin
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Kanai KI, Asano K, Hisamitsu T, Suzaki H. Suppressive activity of epinastine hydrochloride on TARC production from human peripheral blood CD4+ T cells in-vitro. J Pharm Pharmacol 2006; 57:1027-36. [PMID: 16102259 DOI: 10.1211/0022357056640] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Thymus- and activation-regulated chemokine (TARC) is an important molecule in the development and maintenance of allergic diseases. However, there is little information about the influence of anti-allergic agents on TARC production. The aim of this study is to examine the influence of epinastine hydrochloride, an H1-receptor antagonist, on TARC production from human peripheral blood CD4+ T cells using an in-vitro cell culture technique. CD4+ T cells prepared from healthy subjects were cultured in wells coated with a combination of OKT3 and anti-CD28 monoclonal antibody in the presence or absence of epinastine HCl for 24 h. The cells were also stimulated with interleukin (IL)-4 in a similar manner. Levels of TARC and IL-4 in culture supernatants were examined by ELISA. The addition of epinastine HCl exerted a dose-dependent suppressive effect on the production of both TARC and IL-4 from CD4+ T cells under co-stimulatory molecule stimulation. The minimum concentration of the agent showing a significant suppressive effect on TARC and IL-4 production was 5.0 microM and 2.5 microM, respectively. Epinastine HCl also suppressed the ability of cells to produce TARC in response to IL-4 stimulation, when the agent was added to cell cultures at more than 2.5 microM. It was concluded that this inhibitory action of epinastine HCl may be partially responsible for epinastine's attenuating effect on allergic diseases.
Collapse
Affiliation(s)
- Ken-Ichi Kanai
- Department of Otolaryngology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | | | | | | |
Collapse
|
6
|
Spitaler M, Cantrell DA. Protein kinase C and beyond. Nat Immunol 2004; 5:785-90. [PMID: 15282562 DOI: 10.1038/ni1097] [Citation(s) in RCA: 222] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2004] [Accepted: 06/07/2004] [Indexed: 11/08/2022]
Abstract
Protein kinase C molecules regulate both positive and negative signal transduction pathways essential for the initiation and homeostasis of immune responses. There are multiple isoforms of protein kinase C that are activated differently by calcium and diacylglycerol, and these are activated mainly by antigen receptors in T cells, B cells and mast cells. Additionally, mammals express several other diacylglycerol binding proteins that are linked to a network of key signal transduction pathways that control lymphocyte biology. Diacylglycerol and protein kinase C regulate a broad range of gene transcription programs but also modulate integrins, chemokine responses and antigen receptors, thereby regulating lymphocyte adhesion, migration, differentiation and proliferation.
Collapse
Affiliation(s)
- Martin Spitaler
- School of Life Sciences, Division of Cell Biology & Immunology, University of Dundee, MSI/WTB Complex, Dow Street, Dundee DD1 5EH, UK
| | | |
Collapse
|
7
|
Jeevan A, Yoshimura T, Foster G, McMurray DN. Effect of Mycobacterium bovis BCG vaccination on interleukin-1 beta and RANTES mRNA expression in guinea pig cells exposed to attenuated and virulent mycobacteria. Infect Immun 2002; 70:1245-53. [PMID: 11854207 PMCID: PMC127753 DOI: 10.1128/iai.70.3.1245-1253.2002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The effect of Mycobacterium bovis BCG vaccination on interleukin-1 beta (IL-1 beta) or regulated-upon-activation, normally T-cell-expressed and -secreted chemokine (RANTES) mRNA expression in guinea pig spleen cells stimulated with concanavalin A, lipopolysaccharide (LPS), phorbol myristate acetate (PMA) plus ionomycin, or purified protein derivative (PPD) was studied in vitro. Similarly, peritoneal exudate cell-derived macrophages from naïve and BCG-vaccinated guinea pigs were infected with M. bovis BCG, Mycobacterium avium, the attenuated Mycobacterium tuberculosis H37Ra strain, or virulent strains H37Rv and Erdman of M. tuberculosis. Total RNA was subjected to Northern blot analysis using probes generated from guinea pig IL-1 beta or RANTES cDNA. Although IL-1 beta and RANTES mRNA could be detected in the spleen cells from naïve animals stimulated with LPS or PMA plus ionomycin, the levels were significantly enhanced after BCG vaccination. mRNA expression was also elevated in macrophages infected with live mycobacteria after BCG vaccination. However, macrophages infected with the virulent H37Rv strain of M. tuberculosis showed 75 to 90% reductions in IL-1 beta expression and 25 to 60% reductions in RANTES mRNA expression compared with macrophages infected with the attenuated H37Ra strain. The IL-1 beta mRNA levels peaked as soon as 1 h after PPD stimulation and 4 h after M. tuberculosis H37Rv infection of macrophages. In contrast, RANTES mRNA expression was delayed until 48 h after infection. These results indicate that molecular mediators produced in response to various stimuli associated with protective immunity against mycobacteria are upregulated after BCG vaccination; however, a significantly weaker response was observed with virulent M. tuberculosis. These initial studies indicate that BCG vaccination has a positive effect on IL-1 beta and RANTES mRNA expression by host cells in a highly relevant animal tuberculosis model.
Collapse
Affiliation(s)
- Amminikutty Jeevan
- Department of Medical Microbiology and Immunology, Texas A&M University System Health Science Center, College Station, Texas 77843-1114, USA.
| | | | | | | |
Collapse
|
8
|
Ottersbach K, Graham GJ. Aggregation-independent modulation of proteoglycan binding by neutralization of C-terminal acidic residues in the chemokine macrophage inflammatory protein 1alpha. Biochem J 2001; 354:447-53. [PMID: 11171125 PMCID: PMC1221674 DOI: 10.1042/0264-6021:3540447] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Members of the chemokine family of proteins mediate their biological effects through interaction with a family of seven-transmembrane G-protein-coupled receptors. This interaction is complicated by the biochemical properties of chemokines, such as their ability to form self aggregates and their ability to bind to proteoglycans. With some chemokines there is a clear interrelationship between these interactions; the chemokine platelet factor 4 binds preferentially to proteoglycans in its aggregated form. Little is known about the role of aggregation in the proteoglycan binding of other chemokines. Here we demonstrate that the aggregation status of the chemokine macrophage inflammatory protein 1alpha (MIP-1alpha) has no detectable effect on its affinity for proteoglycans. Furthermore, we demonstrate that the alteration of acidic amino acid residues in MIP-1alpha influences the affinity of its interactions with heparin as these residues are progressively neutralized, leading to an enhanced binding affinity for heparin. Thus, with MIP-1alpha, aggregation is not a determinant of proteoglycan binding; however, overall charge does seem to have a major role in the interaction. These results therefore add to our understanding of the nature of the interaction between MIP-1alpha and proteoglycans and suggests that the basic amino acids might not be the sole regulators of proteoglycan binding.
Collapse
Affiliation(s)
- K Ottersbach
- The Beatson Institute for Cancer Research, CRC Beatson Laboratories, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, U.K
| | | |
Collapse
|