1
|
Sun S, Ding Y, Yang D, Shen J, Zhang T, Song G, Chen X, Lin Y, Chen R. Identification of potential hub genes and drugs in septic kidney injury: a bioinformatic analysis with preliminary experimental validation. Front Med (Lausanne) 2025; 12:1502189. [PMID: 40166075 PMCID: PMC11955678 DOI: 10.3389/fmed.2025.1502189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/13/2025] [Indexed: 04/02/2025] Open
Abstract
Background Sepsis-associated kidney injury (SAKI) is a prevalent complication in intensive care unit (ICU) patients with sepsis. Diagnosis currently relies on clinical assessment, urine output, and serum creatinine levels, yet effective clinical treatments remain scarce. Our objectives are to explore prospective, targeted medications for the treatment of septic kidney injury and to employ bioinformatics to identify key genes and pathways that may be implicated in the pathogenesis of SAKI. Methods We utilized the GEO database for differential gene screening. Related genes of septic kidney injury were identified through Pubmed2Ensembl, followed by annotation and visualization of gene ontology biological processes and KEGG pathways using DAVID. Protein-protein interactions were analyzed with the STRING database, and hub genes were identified using Cytoscape software. Candidate genes were further validated through Metascape. The CTD database was employed to uncover the relationship between hub genes and acute kidney injury (AKI). CIBERSORT was applied to evaluate the infiltration of immune cells and their association with hub genes. Hub genes were experimentally verified through qPCR detection. Lastly, the Drug-Gene Interaction Database (DGIdb) was utilized to identify drug-gene interactions. Results Six genes, including TNF, CXCL8, IL-6, IL-1β, IL-2, and IL-10, were associated with three major signaling pathways: the COVID-19 adverse outcome pathway, an overview of pro-inflammatory and pro-fibrotic mediators, and the interleukin-10 signaling pathway. Additionally, 12 targeted drugs were identified as potential therapeutic agents.
Collapse
Affiliation(s)
- Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Dong Yang
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiwei Shen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Guobin Song
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Rui Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Anesthesiology, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
2
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Toll-like receptor response to Zika virus infection: progress toward infection control. NPJ VIRUSES 2025; 3:20. [PMID: 40295746 PMCID: PMC11906774 DOI: 10.1038/s44298-025-00102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025]
Abstract
Infection with the Zika virus (ZIKV) poses a threat to human health. An improved understanding of the host Toll-like receptor response, disease onset, and viral clearance in vivo and in vitro may lead to the development of therapeutic or prophylactic interventions against viral infections. Currently, no clinically approved ZIKV vaccine is available, highlighting the need for its development. In this study, we discuss the progress in the Zika vaccine, including advances in the use of Toll-like receptor agonists as vaccine adjuvants to enhance vaccine efficacy.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, Bangladesh.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
3
|
Sangsuwan R, Thuamsang B, Pacifici N, Tachachartvanich P, Murphy D, Ram A, Albeck J, Lewis JS. Identification of signaling networks associated with lactate modulation of macrophages and dendritic cells. Heliyon 2025; 11:e42098. [PMID: 39975831 PMCID: PMC11835580 DOI: 10.1016/j.heliyon.2025.e42098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
The advancement in the understanding of cancer immune evasion has manifested the development of cancer immunotherapeutic approaches such as checkpoint inhibitors and interleukin agonists. Although cancer immunotherapy breakthroughs have demonstrated improved potency for cancer treatment, only a fraction of patients effectively respond to these treatments. Moreover, there is compelling evidence indicating that cancer cells develop a unique microenvironment through adaptive metabolic reprogramming, which aberrantly modulates host immunity to evade immunosurveillance. As part of the tumor cell adaptive metabolic switch, lactate is produced and released into the tumor environment. Recent studies have shown that lactate significantly modulates immune functions, especially in innate immune cells. Dendritic cells (DCs) and macrophages (MΦs) are specialized antigen-presenting cells serving as key players in innate immunity and anticancer-associated immune responses. Although most studies have shown that lactate affects immune phenotypes (e.g., surface protein expression and cytokine production), the cell signaling network mediated by lactate is not fully understood. In the present study, we identified the key signaling pathways in bone marrow-derived DCs and MΦs that were changed by cancer-relevant concentrations of lactate. First, transcriptome analysis was used to guide notable signaling pathways mediated by lactate. Subsequently, biomolecular techniques, including immunoblotting, flow cytometry, and immunofluorescence imaging were performed to corroborate the changes in these key signaling pathways at the protein level. The results indicated that lactate differentially impacted the biochemical networks of DCs and MΦs. While lactate mainly altered STAT3, ERK, and p38 MAPK signaling cascades in DCs, the STAT1 and GSK-3β signaling in MΦs were the major pathways significantly impacted by lactate. This study identifies key biochemical pathways in innate immune cells that are impacted by lactate, which advances our understanding of the interplay between the tumor microenvironment and innate immunity.
Collapse
Affiliation(s)
- Rapeepat Sangsuwan
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
- Laboratory of Natural Products, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Bhasirie Thuamsang
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - Noah Pacifici
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - Phum Tachachartvanich
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Devan Murphy
- Department of Molecular and Cell Biology, University of California, Davis, CA, 95616, USA
| | - Abhineet Ram
- Department of Molecular and Cell Biology, University of California, Davis, CA, 95616, USA
| | - John Albeck
- Department of Molecular and Cell Biology, University of California, Davis, CA, 95616, USA
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, FL, 32611, USA
| |
Collapse
|
4
|
Gachpazan M, Alashti AA, Jahantigh HR, Moghbeli M, Faezi S, Hosseini SY, Eftekharian MM, Nasimi M, Khiavi FM, Rahimi A, Mianroodi RA, Pakjoo M, Taghizadeh M, Tempesta M, Mahdavi M. Immunization with recombinant HPV16-E7d in fusion with Flagellin as a cancer vaccine: Effect of antigen-adjuvant orientation on the immune response pattern. Immunol Res 2025; 73:50. [PMID: 39939497 DOI: 10.1007/s12026-025-09598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/19/2025] [Indexed: 02/14/2025]
Abstract
Human papillomavirus (HPV) is the leading cause of cervical cancer worldwide. The pathogenesis of HPV is mainly dependent on its E7 and E6 proteins. Up to now, different adjuvants have been used to enhance the efficacy of the immune response against these two proteins. In this study, Flagellin (FLA) was used as adjuvant to test adjuvant activity and also see whether its orientation of attachment can affect the immune response pattern. The E7d-FLA and FLA-E7d in pET28a vector were constructed and then the recombinant proteins were expressed in E. coli BL21 (DE3) bacteria under IPTG induction. The expression of recombinant E7d-FLA and FLA-E7d proteins is confirmed by SDS-PAGE and western blot. Then, recombinant fusion proteins were purified using a nickel-nitrilotriacetic acid (Ni-NTA) column. The recombinant proteins were checked for endotoxin contamination and then quantified by Bradford. Eight-to-ten-week-old male Balb/C mice were immunized subcutaneously with 10 µg recombinant E7d-FLA, FLA-E7d and HPV16E7d vaccine on days 0, 14 and 28. In addition, PBS and FLA groups were considered as control group. Then, spleen cells were harvested to assess lymphocyte proliferation and IFN-γ, IL-4 and IL-17 cytokines. In addition, mice sera were used for specific total IgG and IgG1, IgG2a, IgG2b and IgM antibodies assessment by ELISA. The results show that E7d-FLA is more potent in the induction of lymphocyte proliferation, CTL response and specific total IgG, IgG2a and IgG2b response, while the FLA-E7d vaccine was associated with more IFN-γ, and IL-17 cytokine response. The results of this study proved the ability of FLA as an adjuvant in fusion with E7d in the induction of cellular and humoral immune responses. In addition, it also emphasizes that antigen-adjuvant orientation can affect the immune response strength and polarization against HPV E7d vaccine candidate. HIGHLIGHTS: Flagellin is attached to HPV-16 E7d at the C- or N-terminus to create E7d-FLA and FLA-E7d candidate vaccines. The E7d-FLA vaccine showed a significant increase in lymphocyte proliferation, CTL response and IgG response versus FLA-E7d vaccine. The FLA-E7d vaccine is associated with a significant increase in IFN-γ and IL-17 cytokines response versus E7d-FLA vaccine. It seems that that antigen-adjuvant orientation is an important parameter in the strength and polarization of immune response in HPV E7d vaccine candidate.
Collapse
Affiliation(s)
- Meysam Gachpazan
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran
- Department of Biology, Islamic Azad University of Damghan Branch, Damghan, Iran
| | - Ali Ahmadnia Alashti
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran
| | - Hamid Reza Jahantigh
- Department of Pathology, Faculty of Medicine, Emory University, Atlanta, GA, 30033, USA
- Interdisciplinary Department of Medicine - Section of Occupational Medicine, University of Bari, Bari, Italy
| | - Majid Moghbeli
- Department of Biology, Islamic Azad University of Damghan Branch, Damghan, Iran
| | - Sobhan Faezi
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Nasimi
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Motavalli Khiavi
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Etemad Zadeh Street, Fatemi-Gharbi Street, Tehran, Iran
| | - Alireza Rahimi
- Department of Recombinant Products, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Arabi Mianroodi
- Department of Research and Development, Research and Production Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Pakjoo
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran
| | - Morteza Taghizadeh
- Department of Medical Vaccine, Agricultural Research, Education and Extension Organization (AREEO), Razi Vaccine and Serum Research Institute, Karaj, Iran.
| | - Maria Tempesta
- Department of Veterinary Medicine, Animal Health and Zoonosis PhD Course, University of Bari, Bari, Italy
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran.
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran.
| |
Collapse
|
5
|
Hamze Sinno S, Imperatore JA, Bai S, Gomes-Jourdan N, Mafarachisi N, Coronnello C, Zhang L, Jašarević E, Osmanbeyoglu HU, Buckanovich RJ, Cascio S. Egfl6 promotes ovarian cancer progression by enhancing the immunosuppressive functions of tumor-associated myeloid cells. J Clin Invest 2024; 134:e175147. [PMID: 39312740 PMCID: PMC11527450 DOI: 10.1172/jci175147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/11/2024] [Indexed: 09/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) play a critical role in resistance to immunotherapy. In this study, we identified epidermal growth factor-like 6 (Egfl6) as a regulator of myeloid cell functions. Our analyses indicated that Egfl6, via binding with β3 integrins and activation of p38 and SYK signaling, acts as a chemotactic factor for myeloid cell migration and promotes their differentiation toward an immunosuppressive state. In syngeneic mouse models of ovarian cancer (OvCa), tumor expression of Egfl6 increased the intratumoral accumulation of polymorphonuclear (PMN) MDSCs and TAMs and their expression of immunosuppressive factors, including CXCL2, IL-10, and PD-L1. Consistent with this, in an immune 'hot' tumor model, Egfl6 expression eliminated response to anti-PD-L1 therapy, while Egfl6 neutralizing antibody decreased the accumulation of tumor-infiltrating CD206+ TAMs and PMN-MDSCs and restored the efficacy of anti-PD-L1 therapy. Supporting a role in human tumors, in human OvCa tissue samples, areas of high EGFL6 expression colocalized with myeloid cell infiltration. scRNA-Seq analyses revealed a correlation between EGFL6 and immune cell expression of immunosuppressive factors. Our data provide mechanistic insights into the oncoimmunologic functions of EGFL6 in mediating tumor immune suppression and identified EGFL6 as a potential therapeutic target to enhance immunotherapy in patients with OvCa.
Collapse
Affiliation(s)
- Sarah Hamze Sinno
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Shoumei Bai
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | - Linan Zhang
- Department of Applied Mathematics, School of Mathematics and Statistics, Ningbo University, Ningbo, Zhejiang, China
| | - Eldin Jašarević
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Computational and Systems Biology, Pittsburgh, Pennsylvania, USA
| | - Hatice U. Osmanbeyoglu
- Department of Biomedical Informatics, School of Medicine
- UPMC Hillman Cancer Center
- Department of Bioengineering, School of Engineering, and
| | - Ronald J. Buckanovich
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sandra Cascio
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center
| |
Collapse
|
6
|
Xuan S, Ma Y, Zhou H, Gu S, Yao X, Zeng X. The implication of dendritic cells in lung diseases: Immunological role of toll-like receptor 4. Genes Dis 2024; 11:101007. [PMID: 39238498 PMCID: PMC11375267 DOI: 10.1016/j.gendis.2023.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 09/07/2024] Open
Abstract
The immune responses play a profound role in the progression of lung lesions in both infectious and non-infectious diseases. Dendritic cells, as the "frontline" immune cells responsible for antigen presentation, set up a bridge between innate and adaptive immunity in the course of these diseases. Among the receptors equipped in dendritic cells, Toll-like receptors are a group of specialized receptors as one type of pattern recognition receptors, capable of sensing environmental signals including invading pathogens and self-antigens. Toll-like receptor 4, a pivotal member of the Toll-like receptor family, was formerly recognized as a receptor sensitive to the outer membrane component lipopolysaccharide derived from Gram-negative bacteria, triggering the subsequent response. Moreover, its other essential roles in immune responses have drawn significant attention in the past decade. A better understanding of the implication of Toll-like receptor 4 in dendritic cells could contribute to the management of pulmonary diseases including pneumonia, pulmonary tuberculosis, asthma, acute lung injury, and lung cancer.
Collapse
Affiliation(s)
- Shurui Xuan
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Yuan Ma
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Honglei Zhou
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Shengwei Gu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xin Yao
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaoning Zeng
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
7
|
Geng J, Chrabaszczewska M, Kurpiejewski K, Stankiewicz-Drogon A, Jankowska-Anyszka M, Darzynkiewicz E, Grzela R. Cap-related modifications of RNA regulate binding to IFIT proteins. RNA (NEW YORK, N.Y.) 2024; 30:1292-1305. [PMID: 39009378 PMCID: PMC11404448 DOI: 10.1261/rna.080011.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
All cells in our body are equipped with receptors to recognize pathogens and trigger a rapid defense response. As a result, foreign molecules are blocked, and cells are alerted to the danger. Among the many molecules produced in response to viral infection are interferon-induced proteins with tetratricopeptide repeats (IFITs). Their role is to recognize foreign mRNA and eliminate it from the translational pool of transcripts. In the present study, we used biophysical methods to characterize the interactions between the IFIT1 protein and its partners IFIT2 and IFIT3. IFIT1 interacts with IFIT3 with nanomolar binding affinity, which did not change significantly in the presence of the preformed IFIT2/3 complex. The interactions between IFIT2 and IFIT3 and IFIT1 and IFIT2 were one order of magnitude weaker. We also present kinetic data of the interactions between the IFIT protein complex and short RNA bearing various modifications at the 5' end. We show kinetic parameters for interaction between the IFIT complex and RNA with m6Am modification. The results show that the cap-adjacent m6Am modification is a stronger signature than cap1 alone. It blocks the formation of a complex between IFIT proteins and m7Gpppm6Am-RNA much more effectively than other cap modifications. In contrast, m6A in the 5'UTR is not recognized by IFIT proteins and does not contribute to translation repression by IFIT proteins. The data obtained are important for understanding the regulation of expression of genetic information. They indicate that 2'-O and m6Am modifications modulate the availability of mRNA molecules for proteins of innate immune response.
Collapse
Affiliation(s)
- Jingping Geng
- Interdisciplinary Laboratory of Molecular Biology and Biophysics, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Magdalena Chrabaszczewska
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | | | - Anna Stankiewicz-Drogon
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | | | - Edward Darzynkiewicz
- Interdisciplinary Laboratory of Molecular Biology and Biophysics, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | - Renata Grzela
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| |
Collapse
|
8
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Recent Insights into the Molecular Mechanisms of the Toll-like Receptor Response to Influenza Virus Infection. Int J Mol Sci 2024; 25:5909. [PMID: 38892096 PMCID: PMC11172706 DOI: 10.3390/ijms25115909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Influenza A viruses (IAVs) pose a significant global threat to human health. A tightly controlled host immune response is critical to avoid any detrimental effects of IAV infection. It is critical to investigate the association between the response of Toll-like receptors (TLRs) and influenza virus. Because TLRs may act as a double-edged sword, a balanced TLR response is critical for the overall benefit of the host. Consequently, a thorough understanding of the TLR response is essential for targeting TLRs as a novel therapeutic and prophylactic intervention. To date, a limited number of studies have assessed TLR and IAV interactions. Therefore, further research on TLR interactions in IAV infection should be conducted to determine their role in host-virus interactions in disease causation or clearance of the virus. Although influenza virus vaccines are available, they have limited efficacy, which should be enhanced to improve their efficacy. In this study, we discuss the current status of our understanding of the TLR response in IAV infection and the strategies adopted by IAVs to avoid TLR-mediated immune surveillance, which may help in devising new therapeutic or preventive strategies. Furthermore, recent advances in the use of TLR agonists as vaccine adjuvants to enhance influenza vaccine efficacy are discussed.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
9
|
Chen C, Chen Y, Lu M, Xu L, Yan R, Li X, Song X. IFN-γ inhibitory molecules derived from Eimeria maxima inhibit IL-12 secretion by modulating MAPK pathways in chicken macrophages. Poult Sci 2024; 103:103359. [PMID: 38128458 PMCID: PMC10776662 DOI: 10.1016/j.psj.2023.103359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023] Open
Abstract
IFN-γ plays a crucial role in resisting intracellular parasitic protozoa, such as Eimeria species. In our previous study, we identified 4 molecules derived from Eimeria maxima (E. maxima) that significantly inhibited IFN-γ production. However, the mechanism underlying this inhibitory effect remains unknown. In this study, we first investigated the effects of these 4 IFN-γ inhibitory molecules on the expression levels of chicken Toll-like receptors (chTLRs), IL-12, IL-10, TGF-β, and TNF-α in chicken macrophage HD11 and bone marrow-derived dendritic cells (BMDCs). The results demonstrated that these 4 inhibitory molecules significantly downregulated the mRNA levels of chTLR-2, chTLR-4, chTLR-21, and both mRNA and protein levels of IL-12. Subsequently, to clarify the effects of these 4 inhibitory molecules on the IL-12 secretion-related signaling pathways in chicken macrophages, qRT-PCR and Western blot were used to detect the changes of key molecules involved in the signaling pathways of IL-12 secretion (NF-κB, ERK1/2, p38, JNK, STAT3) following coincubation with these inhibitory molecules. Finally, RNAi was employed to verify the function of key molecules in the signaling pathway. The results revealed a significant upregulation in the expression of ERK1/2 phosphorylated protein induced by the 4 inhibitory molecules. Knockdown of the ERK1/2 gene significantly reduced the inhibitory effect of the 4 E. maxima inhibitory molecules on IL-12. These findings indicate that the 4 inhibitory molecules can inhibit the secretion of IL-12 by upregulating the expression of ERK1/2 phosphorylated protein, which is a key molecule in the ERK-MAPK pathway. Our study may contribute to elucidating the mechanisms underlying immune evasion during E. maxima infections, thereby providing new insights for the control of chicken coccidiosis.
Collapse
Affiliation(s)
- Chen Chen
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yufeng Chen
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Mingmin Lu
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lixin Xu
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ruofeng Yan
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiangrui Li
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiaokai Song
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
10
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Viral Liver Disease and Intestinal Gut–Liver Axis. GASTROINTESTINAL DISORDERS 2024; 6:64-93. [DOI: 10.3390/gidisord6010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
The intestinal microbiota is closely related to liver diseases via the intestinal barrier and bile secretion to the gut. Impairment of the barrier can translocate microbes or their components to the liver where they can contribute to liver damage and fibrosis. The components of the barrier are discussed in this review along with the other elements of the so-called gut–liver axis. This bidirectional relation has been widely studied in alcoholic and non-alcoholic liver disease. However, the involvement of microbiota in the pathogenesis and treatment of viral liver diseases have not been extensively studied, and controversial data have been published. Therefore, we reviewed data regarding the integrity and function of the intestinal barrier and the changes of the intestinal microbioma that contribute to progression of Hepatitis B (HBV) and Hepatitis C (HCV) infection. Their consequences, such as cirrhosis and hepatic encephalopathy, were also discussed in connection with therapeutic interventions such as the effects of antiviral eradication and the use of probiotics that may influence the outcome of liver disease. Profound alterations of the microbioma with significant reduction in microbial diversity and changes in the abundance of both beneficial and pathogenic bacteria were found.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Ioannis Tsomidis
- Department of Gastroenterology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece
| |
Collapse
|
11
|
Elemam NM, Mekky RY, Rashid G, Braoudaki M, Youness RA. Pharmacogenomic and epigenomic approaches to untangle the enigma of IL-10 blockade in oncology. Expert Rev Mol Med 2024; 26:e1. [PMID: 38186186 PMCID: PMC10941350 DOI: 10.1017/erm.2023.26] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 01/09/2024]
Abstract
The host immune system status remains an unresolved mystery among several malignancies. An immune-compromised state or smart immune-surveillance tactics orchestrated by cancer cells are the primary cause of cancer invasion and metastasis. Taking a closer look at the tumour-immune microenvironment, a complex network and crosstalk between infiltrating immune cells and cancer cells mediated by cytokines, chemokines, exosomal mediators and shed ligands are present. Cytokines such as interleukins can influence all components of the tumour microenvironment (TME), consequently promoting or suppressing tumour invasion based on their secreting source. Interleukin-10 (IL-10) is an interlocked cytokine that has been associated with several types of malignancies and proved to have paradoxical effects. IL-10 has multiple functions on cellular and non-cellular components within the TME. In this review, the authors shed the light on the regulatory role of IL-10 in the TME of several malignant contexts. Moreover, detailed epigenomic and pharmacogenomic approaches for the regulation of IL-10 were presented and discussed.
Collapse
Affiliation(s)
- Noha M. Elemam
- Research Instiute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Radwa Y. Mekky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo 12622, Egypt
| | - Gowhar Rashid
- Amity Medical School, Amity University, Gurugram (Manesar) 122413, Haryana, India
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK
| | - Rana A. Youness
- Biology and Biochemistry Department, Faculty of Biotechnology, German International University, Cairo 11835, Egypt
| |
Collapse
|
12
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. TLR agonists as vaccine adjuvants in the prevention of viral infections: an overview. Front Microbiol 2023; 14:1249718. [PMID: 38179453 PMCID: PMC10764465 DOI: 10.3389/fmicb.2023.1249718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024] Open
Abstract
Tol-like receptor (TLR) agonists, as potent adjuvants, have gained attention in vaccine research for their ability to enhance immune responses. This study focuses on their application in improving vaccine efficacy against key viral infections, including hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), SARS-CoV-2, influenza virus, and flaviviruses, including West Nile virus, dengue virus, and chikungunya virus. Vaccines are crucial in preventing microbial infections, including viruses, and adjuvants play a vital role in modulating immune responses. However, there are still many diseases for which effective vaccines are lacking or have limited immune response, posing significant threats to human health. The use of TLR agonists as adjuvants in viral vaccine formulations holds promise in improving vaccine effectiveness. By tailoring adjuvants to specific pathogens, such as HBV, HCV, HIV, SARS-CoV-2, influenza virus, and flavivirus, protective immunity against chronic and emerging infectious disease can be elicited.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
13
|
Khalaf JK, Bess LS, Walsh LM, Ward JM, Johnson CL, Livesay MT, Jackson KJ, Evans JT, Ryter KT, Bazin-Lee HG. Diamino Allose Phosphates: Novel, Potent, and Highly Stable Toll-like Receptor 4 Agonists. J Med Chem 2023; 66:13900-13917. [PMID: 37847244 DOI: 10.1021/acs.jmedchem.3c00724] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Most known synthetic toll-like receptor 4 (TLR4) agonists are carbohydrate-based lipid-A mimetics containing several fatty acyl chains, including a labile 3-O-acyl chain linked to the C-3 position of the non-reducing sugar known to undergo cleavage impacting stability and resulting in loss of activity. To overcome this inherent instability, we rationally designed a new class of chemically more stable synthetic TLR4 ligands that elicit robust innate and adaptive immune responses. This new class utilized a diamino allose phosphate (DAP) scaffold containing a nonhydrolyzable 3-amide bond instead of the classical 3-ester. Accordingly, the DAPs have significantly improved thermostability in aqueous formulations and potency relative to other known natural and synthetic TLR4 ligands. Furthermore, the DAP analogues function as potent vaccine adjuvants to enhance influenza-specific antibodies in mice and provide protection against lethal influenza virus challenges. This novel set of TLR4 ligands show promise as next-generation vaccine adjuvants and stand-alone immunomodulators.
Collapse
Affiliation(s)
- Juhienah K Khalaf
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Laura S Bess
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Lois M Walsh
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Janine M Ward
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Craig L Johnson
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Mark T Livesay
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Konner J Jackson
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Jay T Evans
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Kendal T Ryter
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| | - Hélène G Bazin-Lee
- Inimmune Corporation, 1121 E Broadway, Suite 121, Missoula, Montana 59802, United States
| |
Collapse
|
14
|
Mazzarino M, Cetin E, Bartosova M, Marinovic I, Ipseiz N, Hughes TR, Schmitt CP, Ramji DP, Labéta MO, Raby AC. Therapeutic targeting of chronic kidney disease-associated DAMPs differentially contributing to vascular pathology. Front Immunol 2023; 14:1240679. [PMID: 37849759 PMCID: PMC10577224 DOI: 10.3389/fimmu.2023.1240679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/08/2023] [Indexed: 10/19/2023] Open
Abstract
Chronic Kidney Disease (CKD) is associated with markedly increased cardiovascular (CV) morbidity and mortality. Chronic inflammation, a hallmark of both CKD and CV diseases (CVD), is believed to drive this association. Pro-inflammatory endogenous TLR agonists, Damage-Associated Molecular Patterns (DAMPs), have been found elevated in CKD patients' plasma and suggested to promote CVD, however, confirmation of their involvement, the underlying mechanism(s), the extent to which individual DAMPs contribute to vascular pathology in CKD and the evaluation of potential therapeutic strategies, have remained largely undescribed. A multi-TLR inhibitor, soluble TLR2, abrogated chronic vascular inflammatory responses and the increased aortic atherosclerosis-associated gene expression observed in nephropathic mice, without compromising infection clearance. Mechanistically, we confirmed elevation of 4 TLR DAMPs in CKD patients' plasma, namely Hsp70, Hyaluronic acid, HMGB-1 and Calprotectin, which displayed different abilities to promote key cellular responses associated with vascular inflammation and progression of atherosclerosis in a TLR-dependent manner. These included loss of trans-endothelial resistance, enhanced monocyte migration, increased cytokine production, and foam cell formation by macrophages, the latter via cholesterol efflux inhibition. Calprotectin and Hsp70 most consistently affected these functions. Calprotectin was further elevated in CVD-diagnosed CKD patients and strongly correlated with the predictor of CV events CRP. In nephropathic mice, Calprotectin blockade robustly reduced vascular chronic inflammatory responses and pro-atherosclerotic gene expression in the blood and aorta. Taken together, these findings demonstrated the critical extent to which the DAMP-TLR pathway contributes to vascular inflammatory and atherogenic responses in CKD, revealed the mechanistic contribution of specific DAMPs and described two alternatives therapeutic approaches to reduce chronic vascular inflammation and lower CV pathology in CKD.
Collapse
Affiliation(s)
- Morgane Mazzarino
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Esra Cetin
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Iva Marinovic
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Natacha Ipseiz
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Timothy R. Hughes
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Dipak P. Ramji
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Mario O. Labéta
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Anne-Catherine Raby
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
15
|
Chu YT, Liao MT, Tsai KW, Lu KC, Hu WC. Interplay of Chemokines Receptors, Toll-like Receptors, and Host Immunological Pathways. Biomedicines 2023; 11:2384. [PMID: 37760825 PMCID: PMC10525553 DOI: 10.3390/biomedicines11092384] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
A comprehensive framework has been established for understanding immunological pathways, which can be categorized into eradicated and tolerable immune responses. Toll-like receptors (TLRs) are associated with specific immune responses. TH1 immunity is related to TLR7, TLR8, and TLR9, while TH2 immunity is associated with TLR1, TLR2, and TLR6. TH22 immunity is linked to TLR2, TLR4, and TLR5, and THαβ (Tr1) immunity is related to TLR3, TLR7, and TLR9. The chemokine receptor CXCR5 is a marker of follicular helper T cells, and other chemokine receptors can also be classified within a framework based on host immunological pathways. On the basis of a literature review on chemokines and immunological pathways, the following associations were identified: CCR5 with TH1 responses, CCR1 with TH1-like responses, CCR4 (basophils) and CCR3 (eosinophils) with TH2 and TH9 responses, CCR10 with TH22 responses, CCR6 with TH17 responses, CXCR3 with THαβ responses, CCR8 with regulatory T cells (Treg), and CCR2 with TH3 responses. These findings contribute to the identification of biomarkers for immune cells and provide insights into host immunological pathways. Understanding the chemokine and Toll-like receptor system is crucial for comprehending the function of the innate immune system, as well as adaptive immune responses.
Collapse
Affiliation(s)
- Yuan-Tung Chu
- Department of Anatomic Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu 300, Taiwan;
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Kuo-Wang Tsai
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (K.-W.T.); (K.-C.L.)
| | - Kuo-Cheng Lu
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (K.-W.T.); (K.-C.L.)
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
| | - Wan-Chung Hu
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (K.-W.T.); (K.-C.L.)
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Department of Biotechnology, Ming Chuan University, Taoyuan 333, Taiwan
| |
Collapse
|
16
|
Xu Q, Sheng L, Zhu X, Liu Z, Wei G, Zhang T, Du H, Yang A, Yao J, Zhang G, Sun R. Jingfang granules exert anti-psoriasis effect by targeting MAPK-mediated dendritic cell maturation and PPARγ-mediated keratinocytes cell cycle progression in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154925. [PMID: 37321079 DOI: 10.1016/j.phymed.2023.154925] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Jingfang granules (JFG), derived from JingFangBaiDu San (JFBDS), are a traditional herbal formulas used for the treatment of respiratory tract infections. They were initially prescribed to treat skin disease, such as psoriasis in Chinese Taiwan, but are not widely used for psoriasis treatment in mainland China because of the lack of anti-psoriasis mechanism research. PURPOSES The present study was designed to evaluate the anti-psoriasis effect of JFG and reveal the correlated mechanisms of JFG in vivo and in vitro using network pharmacology, UPLC-Q-TOF-MS technology and molecular biotechnology methods. RESULTS An imiquimod-induced psoriasis-like murine model was used to verify the anti-psoriasis effect in vivo, with inhibition of lymphocytosis and CD3+CD19+B cell proliferation in the peripheral blood and prevention of the activation of CD4+IL17+T cells and CD11c+ MHC Ⅱ+ dendritic cells (DCs) in the spleen. Network pharmacology analysis demonstrated that the targets of the active components were significantly enriched in pathways involved in cancer, inflammatory bowel disease and rheumatoid arthritis, which were closely related to cell proliferation and immune regulation. The drug-component-target networks and molecular docking analysis demonstrated the active ingredients to be luteolin, naringin and 6'-feruloylnodakenin, which had a good binding affinity to PPARγ, p38a MAPK and TNF-a. Finally, UPLC-Q-TOF-MS analysis to validate the active ingredients in drug-containing serum and in vitro experiments showed that JFG inhibited the maturation and activation of BMDCs via the p38a MAPK signaling pathway and translocation of the agonist PPARγ into the nuclei to reduce the activity of NF-κB/STAT3 inflammatory signaling pathway in keratinocytes. CONCLUSIONS Our study demonstrated that JFG improved psoriasis by inhibiting the maturation and activation of BMDCs and proliferation and inflammation of keratinocytes, which may facilitate the applications of JFG in anti-psoriasis therapy in clinical settings.
Collapse
Affiliation(s)
- Qingqing Xu
- Department of Dermato-Venereology, The Second Hospital of Shandong University, Jinan 250033, China; State Key laboratory of Generic Manufacture Technology of Chines Traditional Medicine, Lunan Pharmaceutical Co., Ltd., Linyi 276005, China; Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lisong Sheng
- State Key laboratory of Generic Manufacture Technology of Chines Traditional Medicine, Lunan Pharmaceutical Co., Ltd., Linyi 276005, China; Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Xia Zhu
- Department of Dermato-Venereology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Zhaoyang Liu
- Department of Dermato-Venereology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Guo Wei
- Department of Dermato-Venereology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Tianyu Zhang
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Hang Du
- The Second Hospital of Shandong University, Jinan 250033, China
| | - Anbo Yang
- Department of Dermato-Venereology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Jingchun Yao
- State Key laboratory of Generic Manufacture Technology of Chines Traditional Medicine, Lunan Pharmaceutical Co., Ltd., Linyi 276005, China; Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi 273400, China
| | - Guimin Zhang
- State Key laboratory of Generic Manufacture Technology of Chines Traditional Medicine, Lunan Pharmaceutical Co., Ltd., Linyi 276005, China; Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi 273400, China.
| | - Rong Sun
- The Second Hospital of Shandong University, Jinan 250033, China; Advanced Medical Research Institute, Shandong University, Jinan 250012, China.
| |
Collapse
|
17
|
Bernal AM, Sosa FN, Todero MF, Montagna DR, Vermeulen ME, Fernández-Brando RJ, Ramos MV, Errea AJ, Rumbo M, Palermo MS. Nasal immunization with H7 flagellin protects mice against hemolytic uremic syndrome secondary to Escherichia coli O157:H7 gastrointestinal infection. Front Cell Infect Microbiol 2023; 13:1143918. [PMID: 37260706 PMCID: PMC10227447 DOI: 10.3389/fcimb.2023.1143918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/26/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Shiga-toxin (Stx) producing Escherichia coli (STEC) O157:H7 is the most frequent serotype associated with hemolytic uremic syndrome (HUS) after gastrointestinal infections. Protection against HUS secondary to STEC infections has been experimentally assayed through the generation of different vaccine formulations. With focus on patients, the strategies have been mainly oriented to inhibit production of Stx or its neutralization. However, few approaches have been intended to block gastrointestinal phase of this disease, which is considered the first step in the pathogenic cascade of HUS. The aim of this work was to assay H7 flagellin as a mucosal vaccine candidate to prevent the systemic complications secondary to E. coli O157:H7 infections. Materials and methods The cellular and humoral immune response after H7 nasal immunization in mice were studied by the analysis of systemic and intestinal specific antibody production, as well as cytokine production and lymphocyte proliferation against H7 flagellin ex vivo. Results Immunized mice developed a strong and specific anti-H7 IgG and IgA response, at systemic and mucosal level, as well as a cellular Th1/Th2/Th17 response. H7 induced activation of bone marrow derived dendritic cells in vitro and a significant delayed-type hypersensitivity (DTH) response in immunized mice. Most relevant, immunized mice were completely protected against the challenge with an E. coli O157:H7 virulent strain in vivo, and surviving mice presented high titres of anti-H7 and Stx antibodies. Discussion These results suggest that immunization avoids HUS outcome and allows to elicit a specific immune response against other virulence factors.
Collapse
Affiliation(s)
- Alan Mauro Bernal
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Fernando Nicolás Sosa
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Florencia Todero
- Laboratorio de Fisiología de Procesos Inflamatorios, IMEX CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Daniela Romina Montagna
- Laboratorio de Oncología Experimental, IMEX CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Mónica Elba Vermeulen
- Laboratorio de Células Presentadoras de Antígenos y Respuesta Inflamatoria, IMEX CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Romina Jimena Fernández-Brando
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Victoria Ramos
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Agustina Juliana Errea
- Instituto de Estudios Inmunológicos y Fisiopatológicos - CONICET - Universidad Nacional de La Plata, La Plata, Argentina
| | - Martin Rumbo
- Instituto de Estudios Inmunológicos y Fisiopatológicos - CONICET - Universidad Nacional de La Plata, La Plata, Argentina
| | - Marina Sandra Palermo
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Academia Nacional de Medicina, Buenos Aires, Argentina
| |
Collapse
|
18
|
Castrodeza-Sanz J, Sanz-Muñoz I, Eiros JM. Adjuvants for COVID-19 Vaccines. Vaccines (Basel) 2023; 11:vaccines11050902. [PMID: 37243006 DOI: 10.3390/vaccines11050902] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
In recent decades, the improvement of traditional vaccines has meant that we have moved from inactivated whole virus vaccines, which provoke a moderate immune response but notable adverse effects, to much more processed vaccines such as protein subunit vaccines, which despite being less immunogenic have better tolerability profiles. This reduction in immunogenicity is detrimental to the prevention of people at risk. For this reason, adjuvants are a good solution to improve the immunogenicity of this type of vaccine, with much better tolerability profiles and a low prevalence of side effects. During the COVID-19 pandemic, vaccination focused on mRNA-type and viral vector vaccines. However, during the years 2022 and 2023, the first protein-based vaccines began to be approved. Adjuvanted vaccines are capable of inducing potent responses, not only humoral but also cellular, in populations whose immune systems are weak or do not respond properly, such as the elderly. Therefore, this type of vaccine should complete the portfolio of existing vaccines, and could help to complete vaccination against COVID-19 worldwide now and over the coming years. In this review we analyze the advantages and disadvantages of adjuvants, as well as their use in current and future vaccines against COVID-19.
Collapse
Affiliation(s)
- Javier Castrodeza-Sanz
- National Influenza Centre, 47005 Valladolid, Spain
- Preventive Medicine and Public Health Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Iván Sanz-Muñoz
- National Influenza Centre, 47005 Valladolid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León, ICSCYL, 42002 Soria, Spain
| | - Jose M Eiros
- National Influenza Centre, 47005 Valladolid, Spain
- Microbiology Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
- Microbiology Unit, Hospital Universitario Río Hortega, 47013 Valladolid, Spain
| |
Collapse
|
19
|
Wu N, Li X, Ma H, Zhang X, Liu B, Wang Y, Zheng Q, Fan X. The role of the gut microbiota and fecal microbiota transplantation in neuroimmune diseases. Front Neurol 2023; 14:1108738. [PMID: 36816570 PMCID: PMC9929158 DOI: 10.3389/fneur.2023.1108738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota plays a key role in the function of the host immune system and neuroimmune diseases. Alterations in the composition of the gut microbiota can lead to pathology and altered formation of microbiota-derived components and metabolites. A series of neuroimmune diseases, such as myasthenia gravis (MG), multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSDs), Guillain-Barré syndrome (GBS), and autoimmune encephalitis (AIE), are associated with changes in the gut microbiota. Microecological therapy by improving the gut microbiota is expected to be an effective measure for treating and preventing some neuroimmune diseases. This article reviews the research progress related to the roles of gut microbiota and fecal microbiota transplantation (FMT) in neuroimmune diseases.
Collapse
Affiliation(s)
- Nan Wu
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Xizhi Li
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - He Ma
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Xue Zhang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Bin Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Yuan Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,*Correspondence: Yuan Wang ✉
| | - Qi Zheng
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,Qi Zheng ✉
| | - Xueli Fan
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China,Xueli Fan ✉
| |
Collapse
|
20
|
Tsubokawa D. Immunomodulators secreted from parasitic helminths act on pattern recognition receptors. FRONTIERS IN PARASITOLOGY 2023; 1:1091596. [PMID: 39816467 PMCID: PMC11731691 DOI: 10.3389/fpara.2022.1091596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2025]
Abstract
Excretory-secretory (ES) products from parasitic helminths contain immunomodulatory molecules, which can regulate host immune responses. These immunomodulatory molecules are crucial for successful parasitism, and play roles in tissue migration, maturation, and reproduction. Some target pattern recognition receptors (PRRs), including toll-like receptor, C-type lectin receptor, receptor for advanced glycation end products, and nucleotide-binding oligomerization domain-like receptor. PRRs trigger activation of signaling cascades, inducing innate inflammatory responses and adaptive immunity in hosts. This article reviews ES immunomodulators identified in parasitic helminths that act on PRRs, and their PRR-facilitated immune-regulatory mechanisms. In addition, we describe the therapeutic potential of ES immunomodulators for allergic and inflammatory diseases.
Collapse
Affiliation(s)
- Daigo Tsubokawa
- Department of Parasitology and Tropical Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
21
|
Clemen R, Arlt K, Miebach L, von Woedtke T, Bekeschus S. Oxidized Proteins Differentially Affect Maturation and Activation of Human Monocyte-Derived Cells. Cells 2022; 11:cells11223659. [PMID: 36429087 PMCID: PMC9688260 DOI: 10.3390/cells11223659] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
In cancer, antigen-presenting cells (APC), including dendritic cells (DCs), take up and process proteins to mount adaptive antitumor immune responses. This often happens in the context of inflamed cancer, where reactive oxygen species (ROS) are ubiquitous to modify proteins. However, the inflammatory consequences of oxidized protein uptake in DCs are understudied. To this end, we investigated human monocyte-derived cell surface marker expression and cytokine release profiles when exposed to oxidized and native proteins. Seventeen proteins were analyzed, including viral proteins (e.g., CMV and HBV), inflammation-related proteins (e.g., HO1 and HMGB1), matrix proteins (e.g., Vim and Coll), and vastly in the laboratory used proteins (e.g., BSA and Ova). The multifaceted nature of inflammation-associated ROS was mimicked using gas plasma technology, generating reactive species cocktails for protein oxidation. Fourteen oxidized proteins led to elevated surface marker expression levels of CD25, CD40, CD80, CD86, and MHC-II as well as strongly modified release of IL6, IL8, IL10, IL12, IL23, MCP-1, and TNFα compared to their native counterparts. Especially IL8, heme oxygenase 2, and vimentin oxidation gave pronounced effects. Furthermore, protein kinase phospho-array studies in monocyte-derived cells pulsed with native vs. oxidized IL8 and insulin showed enhanced AKT and RSK2 phosphorylation. In summary, our data provide for the first time an overview of the functional consequences of oxidized protein uptake by human monocyte-derived cells and could therefore be a starting point for exploiting such principle in anticancer therapy in the future.
Collapse
Affiliation(s)
- Ramona Clemen
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Kevin Arlt
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Lea Miebach
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Department of General, Thoracic, Vascular, and Visceral Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Thomas von Woedtke
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Correspondence:
| |
Collapse
|
22
|
Sipka AS, Chandler TL, Weichhart T, Schuberth HJ, Mann S. Inhibition of mTOR in bovine monocyte derived macrophages and dendritic cells provides a potential mechanism for postpartum immune dysfunction in dairy cows. Sci Rep 2022; 12:15084. [PMID: 36064574 PMCID: PMC9445052 DOI: 10.1038/s41598-022-19295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/26/2022] [Indexed: 11/29/2022] Open
Abstract
Dairy cattle experience a profound nutrient deficit postpartum that is associated with immune dysfunction characterized by heightened inflammation and reduced pathogen clearance. The activation of the central nutrient-sensing mTOR pathway is comparatively reduced in leukocytes of early postpartum dairy cows during this time of most pronounced nutrient deficit. We assessed the effect of pharmacological mTOR inhibition (Torin-1, rapamycin) on differentiation of monocyte derived classically (M1) and alternatively (M2) activated macrophages (MPh) and dendritic cells (moDC) from 12 adult dairy cows. Treatment with mTOR inhibitors generated M1 MPh with increased oxidative burst and expression of IL12 subunits but decreased phagocytosis and expression of IL1B, IL6, and IL10. In M2 MPh, treatment inhibited expression of regulatory features (CD163, ARG2, IL10) skewing the cells toward an M1-like phenotype. In moDC, mTOR inhibition increased expression of pro-inflammatory cytokines (IL12A, IL12B, IL1B, IL6) and surface CD80. In co-culture with mixed lymphocytes, mTOR-inhibited moDC exhibited a cytokine profile favoring a Th1 response with increased TNF and IFNG production and decreased IL10 concentrations. We conclude that mTOR inhibition in vitro promoted differentiation of inflammatory macrophages with reduced regulatory features and generation of Th1-favoring dendritic cells. These mechanisms could contribute to immune dysregulation in postpartum dairy cows.
Collapse
Affiliation(s)
- Anja S Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, 231 Farrier Road, Ithaca, NY, 14853, USA.
| | - Tawny L Chandler
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, 231 Farrier Road, Ithaca, NY, 14853, USA
| | - Thomas Weichhart
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090, Vienna, Austria
| | - Hans-Joachim Schuberth
- Institute for Immunology, University of Veterinary Medicine, Buenteweg 2, 30559, Hannover, Germany
| | - Sabine Mann
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, 231 Farrier Road, Ithaca, NY, 14853, USA.
| |
Collapse
|
23
|
Kaur A, Baldwin J, Brar D, Salunke DB, Petrovsky N. Toll-like receptor (TLR) agonists as a driving force behind next-generation vaccine adjuvants and cancer therapeutics. Curr Opin Chem Biol 2022; 70:102172. [PMID: 35785601 DOI: 10.1016/j.cbpa.2022.102172] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 05/18/2022] [Indexed: 01/06/2023]
Abstract
Until recently, the development of new human adjuvants was held back by a poor understanding of their mechanisms of action. The field was revolutionized by the discovery of the toll-like receptors (TLRs), innate immune receptors that directly or indirectly are responsible for detecting pathogen-associated molecular patterns (PAMPs) and respond to them by activating innate and adaptive immune pathways. Hundreds of ligands targeting various TLRs have since been identified and characterized as vaccine adjuvants. This work has important implications not only for the development of vaccines against infectious diseases but also for immuno-therapies against cancer, allergy, Alzheimer's disease, drug addiction and other diseases. Each TLR has its own specific tissue localization and downstream gene signalling pathways, providing researchers the opportunity to precisely tailor adjuvants with specific immune effects. TLR agonists can be combined with other TLR or alternative adjuvants to create combination adjuvants with synergistic or modulatory effects. This review provides an introduction to the various classes of TLR adjuvants and their respective signalling pathways. It provides an overview of recent advancements in the TLR field in the past 2-3 years and discusses criteria for selecting specific TLR adjuvants based on considerations, such as disease mechanisms and correlates of protection, TLR immune biasing capabilities, route of administration, antigen compatibility, new vaccine technology platforms, and age- and species-specific effects.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | | | - Deshkanwar Brar
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, Adelaide 5042, Australia; College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia.
| |
Collapse
|
24
|
Fang Z, Peltz G. An automated multi-modal graph-based pipeline for mouse genetic discovery. Bioinformatics 2022; 38:3385-3394. [PMID: 35608290 PMCID: PMC9992076 DOI: 10.1093/bioinformatics/btac356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/18/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Our ability to identify causative genetic factors for mouse genetic models of human diseases and biomedical traits has been limited by the difficulties associated with identifying true causative factors, which are often obscured by the many false positive genetic associations produced by a GWAS. RESULTS To accelerate the pace of genetic discovery, we developed a graph neural network (GNN)-based automated pipeline (GNNHap) that could rapidly analyze mouse genetic model data and identify high probability causal genetic factors for analyzed traits. After assessing the strength of allelic associations with the strain response pattern; this pipeline analyzes 29M published papers to assess candidate gene-phenotype relationships; and incorporates the information obtained from a protein-protein interaction network and protein sequence features into the analysis. The GNN model produces markedly improved results relative to that of a simple linear neural network. We demonstrate that GNNHap can identify novel causative genetic factors for murine models of diabetes/obesity and for cataract formation, which were validated by the phenotypes appearing in previously analyzed gene knockout mice. The diabetes/obesity results indicate how characterization of the underlying genetic architecture enables new therapies to be discovered and tested by applying 'precision medicine' principles to murine models. AVAILABILITY AND IMPLEMENTATION The GNNHap source code is freely available at https://github.com/zqfang/gnnhap, and the new version of the HBCGM program is available at https://github.com/zqfang/haplomap. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zhuoqing Fang
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gary Peltz
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
25
|
Sipka A, Weichhart T, Mann S. Pharmacological inhibition of the mTOR pathway alters phenotype and cytokine expression in bovine monocyte-derived dendritic cells. Vet Immunol Immunopathol 2022; 249:110441. [PMID: 35597229 DOI: 10.1016/j.vetimm.2022.110441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/05/2022] [Accepted: 05/13/2022] [Indexed: 01/20/2023]
Abstract
Epidemiological studies have long demonstrated the association of nutrient status and immune dysfunction in dairy cows. Postpartum dairy cows experiencing a nutrient deficit show a propensity for increased inflammatory response, decreased pathogen clearance, and increased incidence of infectious disease. Studies in cows and other species show that the nutrient sensing mechanistic target of rapamycin (mTOR) signaling pathway could be one potential causal pathway connecting the deficit in nutrient availability and the heightened inflammatory response. Our objective was to investigate the effects of pharmacological mTOR pathway inhibition on phenotype and cytokine expression of bovine monocyte derived dendritic cells (moDC). We differentiated CD14+ monocytes from dairy cows (n = 14) into moDC in the presence or absence of first- or second-generation mTOR inhibitor rapamycin and PP242 (both 100 nM), respectively. On day seven cells were matured with E. coli lipopolysaccharide (LPS, 100 ng/mL) or left unstimulated to represent naïve moDC. Surface expression of CD14, CD40, CD80, and MHCII was measured via flow cytometry. We measured mRNA expression of IL10, IL12A, IL12B, and TNFα by rt-qPCR, and protein concentrations of IL-10 and TFN-α in cell culture supernatants with a bead-based multiplex assay. Cultures from ten cows successfully developed the moDC phenotype in culture without inhibitors, defined as increased surface expression of CD40, CD80, and MHCII compared with naïve moDC. Only data from these cows were considered for the results on effects of mTOR inhibitors. In naïve and mature moDC mTOR inhibition increased MHCII expression compared to controls. In mature moDC, in addition to MHCII, CD80 expression was increased compared with untreated LPS-stimulated controls. Expression of IL12A mRNA was upregulated in mature, mTOR inhibited moDC compared with untreated controls. In cell culture supernatants mTOR inhibition reduced IL-10 and increased TNF-α concentrations in naïve and mature moDCs compared with untreated controls. Overall rapamycin had a more consistent effect on altering phenotype and cytokine expression of moDC than PP242. In summary we observed an increased expression of co-stimulatory molecules and antigen presentation potential in mature moDC differentiated under mTOR inhibition, and a cytokine pattern that would potentially favor a Th1 type response. This study provides novel data indicating a role for mTOR signaling in bovine moDC phenotype and mediator profile. This proof-of-concept study demonstrates the role of the mTOR pathway in shaping the bovine immune response and may help to provide mechanistic insight and opportunities for modulation of the immune response during the nutrient deficit of early lactation.
Collapse
Affiliation(s)
- Anja Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - Thomas Weichhart
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria
| | - Sabine Mann
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
26
|
Global profiling reveals common and distinct N6-methyladenosine (m6A) regulation of innate immune responses during bacterial and viral infections. Cell Death Dis 2022; 13:234. [PMID: 35288544 PMCID: PMC8921188 DOI: 10.1038/s41419-022-04681-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 12/15/2022]
Abstract
N6-methyladenosine (m6A) is a dynamic post-transcriptional RNA modification influencing all aspects of mRNA biology. While m6A modifications during numerous viral infections have been described, the role of m6A in innate immune response remains unclear. Here, we examined cellular m6A epitranscriptomes during infections of Pseudomonas aeruginosa and herpes simplex virus type 1 (HSV-1), and lipopolysaccharide (LPS) stimulation to identify m6A-regulated innate immune response genes. We showed that a significant portion of cellular genes including many innate immune response genes underwent m6A modifications in 5'UTR and 3'UTR. We identified common and distinct m6A-modified genes under different stimulating conditions. Significantly, the expression of a subset of innate immune response genes was positively correlated with m6A level. Importantly, we identified genes that had significant enrichments of m6A peaks during P. aeruginosa infection following knockdown of m6A "eraser" ALKBH5, confirming the regulation of these genes by m6A and ALKBH5. Among them, we confirmed the association of m6A modification with gene expression in immune response genes TNFAIP3, IFIT1, IFIT2 and IFIH1. Taken together, our results revealed the vital role of m6A in regulating innate immunity against bacterial and viral infections. These works also provided rich resources for the scientific community.
Collapse
|
27
|
Tomalka JA, Suthar MS, Deeks SG, Sekaly RP. Fighting the SARS-CoV-2 pandemic requires a global approach to understanding the heterogeneity of vaccine responses. Nat Immunol 2022; 23:360-370. [PMID: 35210622 DOI: 10.1038/s41590-022-01130-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/05/2022] [Indexed: 11/09/2022]
Abstract
Host genetic and environmental factors including age, biological sex, diet, geographical location, microbiome composition and metabolites converge to influence innate and adaptive immune responses to vaccines. Failure to understand and account for these factors when investigating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine efficacy may impair the development of the next generation of vaccines. Most studies aimed at identifying mechanisms of vaccine-mediated immune protection have focused on adaptive immune responses. It is well established, however, that mobilization of the innate immune response is essential to the development of effective cellular and humoral immunity. A comprehensive understanding of the innate immune response and environmental factors that contribute to the development of broad and durable cellular and humoral immune responses to SARS-CoV-2 and other vaccines requires a holistic and unbiased approach. Along with optimization of the immunogen and vectors, the development of adjuvants based on our evolving understanding of how the innate immune system shapes vaccine responses will be essential. Defining the innate immune mechanisms underlying the establishment of long-lived plasma cells and memory T cells could lead to a universal vaccine for coronaviruses, a key biomedical priority.
Collapse
Affiliation(s)
- Jeffrey A Tomalka
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.,Department of Pediatrics, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven G Deeks
- Department of Medicine, University of California at San Francisco School of Medicine, San Francisco, CA, USA
| | - Rafick Pierre Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA. .,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
28
|
Tretiakova DS, Vodovozova EL. Liposomes as Adjuvants and Vaccine Delivery Systems. BIOCHEMISTRY (MOSCOW) SUPPLEMENT. SERIES A, MEMBRANE AND CELL BIOLOGY 2022; 16:1-20. [PMID: 35194485 PMCID: PMC8853224 DOI: 10.1134/s1990747822020076] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022]
Abstract
The review considers liposomes as systems of substantial interest as adjuvant carriers in vaccinology due to their versatility and maximal biocompatibility. Research and development on the use of liposomes and lipid nanoparticles to create subunit vaccines for the prevention and treatment of infectious diseases has been going on for several decades. In recent years, the area has seen serious progress due to the improvement of the technology of industrial production of various high-grade lipids suitable for parenteral administration and the emergence of new technologies and equipment for the production of liposomal preparations. When developing vaccines, it is necessary to take into account how the body’s immune system (innate and adaptive immunity) functions. The review briefly describes some of the fundamental mechanisms underlying the mobilization of immunity when encountering an antigen, as well as the influence of liposome carriers on the processes of internalization of antigens by immunocompetent cells and ways of immune response induction. The results of the studies on the interactions of liposomes with antigen-presenting cells in function of the liposome size, charge, and phase state of the bilayer, which depends on the lipid composition, are often contradictory and should be verified in each specific case. The introduction of immunostimulant components into the composition of liposomal vaccine complexes—ligands of the pathogen-associated molecular pattern receptors—permits modulation of the strength and type of the immune response. The review briefly discusses liposome-based vaccines approved for use in the clinic for the treatment and prevention of infectious diseases, including mRNA-loaded lipid nanoparticles. Examples of liposomal vaccines that undergo various stages of clinical trials are presented.
Collapse
Affiliation(s)
- D S Tretiakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - E L Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
29
|
Kurago Z, Loveless J. Microbial Colonization and Inflammation as Potential Contributors to the Lack of Therapeutic Success in Oral Squamous Cell Carcinoma. FRONTIERS IN ORAL HEALTH 2022; 2:739499. [PMID: 35048056 PMCID: PMC8757816 DOI: 10.3389/froh.2021.739499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
This review discusses the microenvironment of evolving and established conventional oral squamous cell carcinoma, by far the most common oral cancer. The focus of this paper is mainly on the more recent data that describe the role of microorganisms, host-microbial interactions, and in particular, the contributions of cell-surface toll-like receptors on immune system cells and on normal and malignant epithelial cells to their functions that support carcinogenesis. Because carcinomas arising at various host surfaces share much in common, additional information available from studies of other carcinomas is included in the discussion. Accumulating evidence reveals the complex toll-like receptor-mediated tumor-supporting input into many aspects of carcinogenesis via malignant cells, stromal immune cells and non-immune cells, complicating the search for effective treatments.
Collapse
Affiliation(s)
- Zoya Kurago
- Augusta University Dental College of Georgia, Augusta, GA, United States.,Medical College of Georgia, Augusta, GA, United States.,Georgia Cancer Center, Augusta, GA, United States
| | - Jenni Loveless
- Augusta University Dental College of Georgia, Augusta, GA, United States
| |
Collapse
|
30
|
Zamora V, Carlos Andreu-Ballester J, Rodero M, Cuéllar C. Anisakis simplex: Immunomodulatory effects of larval antigens on the activation of Toll like Receptors. Int Immunopharmacol 2021; 100:108120. [PMID: 34537480 DOI: 10.1016/j.intimp.2021.108120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022]
Abstract
AIMS The objective of this investigation is to evaluate the mechanisms Anisakis simplex employs to modify its host immune system, regarding the larval antigens interactions with Toll-Like-Receptors (TLRs). METHODS AND RESULTS In a previous study, we described that the stimulation of bone marrow derived dendritic cells (BMDCs) with A. simplex larval antigens drive an acute inflammatory response in BALB/c mice, but a more discrete and longer response in C57BL/6J. Moreover, when A. simplex larval antigens were combined with TLR agonists (TLR 1/2-9), they modified mainly TLR2, TLR4 and TLR9 agonists responses in both mice strains, and also TLR3, TLR5 and TLR7 in BALB/c. Antigen-presenting ability was analyzed by the detection of CD11c + cells expressing surface markers (CD80-86, MHC I-II), intracellular cytokines (IL-10, IL-12, TNF-α) and intracellular proteins (Myd88, NF-κβ) by Flow Cytometry. Secreted IL-10 was measured by ELISA. CONCLUSION Our findings confirm not only that the host genetic basis plays a role in the development of a Th2/Th1/Treg response, but also it states A. simplex larval antigens present specific mechanisms to modify the innate response of the host. As allergies share common pathways with the immune response against this particular helminth, our results provide a better understanding into the specific mechanisms of A. simplex allergy related diseases.
Collapse
Affiliation(s)
- Vega Zamora
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Spain.
| | | | - Marta Rodero
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| | - Carmen Cuéllar
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| |
Collapse
|
31
|
Francisco S, Arranz A, Merino J, Punzón C, Perona R, Fresno M. Early p38 Activation Regulated by MKP-1 Is Determinant for High Levels of IL-10 Expression Through TLR2 Activation. Front Immunol 2021; 12:660065. [PMID: 34234775 PMCID: PMC8256158 DOI: 10.3389/fimmu.2021.660065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/07/2021] [Indexed: 01/09/2023] Open
Abstract
Toll-like receptors (TLRs) play a crucial role in the recognition of pathogen-derived components as a first line of defense against infections. It has been suggested that depending on the nature of the pathogens, TLRs activation induce a distinct cytokine profile that may contribute to the polarization of the acquired immune response. Here, we investigated the early MAPK signaling activation via TLR4 and TLR2 receptors and its impact in differential cytokine profile by macrophages. We found that TLR2 ligands activated MAPKs p38 and ERK earlier compared to the TLR4 ligand LPS in macrophages. Higher IL-10/IL-12 and IL-10/TNF-α ratios were also observed at later time points in response to TLR2 ligands compared to LPS. The results also indicate an earlier activation of the phosphatase MKP-1 and that MKP-1 KO macrophages show a prolongation in p38 phosphorylation in response to TLR2 stimulation. Furthermore, p38 is critical for IL-10 expression in response to TLR2 ligands, which triggers the macrophage change to a M2 and regulatory phenotype in contrast to the M1 phenotype induced by TLR4 activation. Therefore, the early TLR2-mediated p38 induction contributes for the high IL-10 production, likely as a virulence strategy to suppress host Th1 response against certain types of pathogens.
Collapse
Affiliation(s)
- Sara Francisco
- DIOMUNE S. L., Parque Científico de Madrid, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - Alicia Arranz
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Merino
- DIOMUNE S. L., Parque Científico de Madrid, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Punzón
- DIOMUNE S. L., Parque Científico de Madrid, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rosario Perona
- Instituto de Investigaciones Biomedicas, Alberto Sols Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Fresno
- DIOMUNE S. L., Parque Científico de Madrid, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
32
|
Verma M, Michalec L, Sripada A, McKay J, Sirohi K, Verma D, Sheth D, Martin R, Dyjack N, Seibold MA, Knapp JR, Tu TH, O'Connor BP, Gorska MM, Alam R. The molecular and epigenetic mechanisms of innate lymphoid cell (ILC) memory and its relevance for asthma. J Exp Med 2021; 218:212204. [PMID: 34076685 PMCID: PMC8176441 DOI: 10.1084/jem.20201354] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/11/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Repetitive exposure of Rag1−/− mice to the Alternaria allergen extract generated a form of memory that elicited an asthma-like response upon a subthreshold recall challenge 3–15 wk later. This memory was associated with lung ICOS+ST2+ ILC2s. Genetic, pharmacologic, and antibody-mediated inhibition and adoptive transfer established an essential role for ILC2s in memory-driven asthma. ATAC-seq demonstrated a distinct epigenetic landscape of memory ILC2s and identified Bach2 and AP1 (JunD and Fosl2) motifs as major drivers of altered gene accessibility. scRNA-seq, gene knockout, and signaling studies suggest that repetitive allergenic stress induces a gene repression program involving Nr4a2, Zeb1, Bach2, and JunD and a preparedness program involving Fhl2, FosB, Stat6, Srebf2, and MPP7 in memory ILC2s. A mutually regulated balance between these two programs establishes and maintains memory. The preparedness program (e.g., Fhl2) can be activated with a subthreshold cognate stimulation, which down-regulates repressors and activates effector pathways to elicit the memory-driven phenotype.
Collapse
Affiliation(s)
- Mukesh Verma
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO
| | - Lidia Michalec
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO
| | - Anand Sripada
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO
| | - Jerome McKay
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO
| | - Kapil Sirohi
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO
| | - Divya Verma
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO
| | - Dipa Sheth
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO
| | - Richard Martin
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO.,Department of Pediatrics, National Jewish Health, Denver, CO
| | - Nathan Dyjack
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO
| | - Max A Seibold
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO.,Department of Pediatrics, National Jewish Health, Denver, CO
| | - Jennifer R Knapp
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO
| | - Ting-Hui Tu
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO
| | - Brian P O'Connor
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO
| | - Magdalena M Gorska
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO.,School of Medicine, University of Colorado Denver, Denver, CO
| | - Rafeul Alam
- Division of Allergy & Immunology, Department of Medicine, National Jewish Health, Denver, CO.,School of Medicine, University of Colorado Denver, Denver, CO
| |
Collapse
|
33
|
Rajendran KV, Neelakanta G, Sultana H. Sphingomyelinases in a journey to combat arthropod-borne pathogen transmission. FEBS Lett 2021; 595:1622-1638. [PMID: 33960414 DOI: 10.1002/1873-3468.14103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022]
Abstract
Ixodes scapularis ticks feed on humans and other vertebrate hosts and transmit several pathogens of public health concern. Tick saliva is a complex mixture of bioactive proteins, lipids and immunomodulators, such as I. scapularis sphingomyelinase (IsSMase)-like protein, an ortholog of dermonecrotoxin SMase D found in the venom of Loxosceles spp. of spiders. IsSMase modulates the host immune response towards Th2, which suppresses Th1-mediated cytokines to facilitate pathogen transmission. Arboviruses utilize exosomes for their transmission from tick to the vertebrate host, and exosomes derived from tick saliva/salivary glands suppress C-X-C motif chemokine ligand 12 and interleukin-8 immune response(s) in human skin to delay wound healing and repair processes. IsSMase affects also viral replication and exosome biogenesis, thereby inhibiting tick-to-vertebrate host transmission of pathogenic exosomes. In this review, we elaborate on exosomes and their biogenesis as potential candidates for developing novel control measure(s) to combat tick-borne diseases. Such targets could help with the development of an efficient anti-tick vaccine for preventing the transmission of tick-borne pathogens.
Collapse
Affiliation(s)
- Kundave V Rajendran
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA.,Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
34
|
Abstract
CpG Oligonucleotides (ODN) are immunomodulatory synthetic oligonucleotides specifically designed to stimulate Toll-like receptor 9. TLR9 is expressed on human plasmacytoid dendritic cells and B cells and triggers an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. This chapter reviews recent progress in understanding the mechanism of action of CpG ODN and provides an overview of human clinical trial results using CpG ODN to improve vaccines for the prevention/treatment of cancer, allergy, and infectious disease.
Collapse
Affiliation(s)
| | | | - Dennis M Klinman
- National Cancer Institute, NIH, Frederick, MD, USA.
- Leitman Klinman Consulting, Potomac, MD, USA.
| |
Collapse
|
35
|
Toll-Like Receptor 2-Tpl2-Dependent ERK Signaling Drives Inverse Interleukin 12 Regulation in Dendritic Cells and Macrophages. Infect Immun 2020; 89:IAI.00323-20. [PMID: 33077627 DOI: 10.1128/iai.00323-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/14/2020] [Indexed: 01/22/2023] Open
Abstract
This study investigated responses to Toll-like receptor 2 (TLR2)-driven extracellular signal-related kinase (ERK) signaling in dendritic cells (DCs) versus macrophages. TLR2 signaling was induced with Pam3Cys-Ser-Lys4, and the role of ERK signaling was interrogated pharmacologically with MEK1/2 inhibitor U0126 or genetically with bone marrow-derived macrophages or DCs from Tpl2-/- mice. We assessed cytokine production via enzyme-linked immunosorbent assay (ELISA) or V-Plex, and mRNA levels were assessed via reverse transcriptase quantitative PCR (qRT-PCR). In macrophages, blockade of ERK signaling by pharmacologic or genetic approaches inhibited interleukin 10 (IL-10) expression and increased expression of the p40 subunit shared by IL-12 and IL-23 (IL-12/23p40). In DCs, blockade of ERK signaling similarly inhibited IL-10 expression but decreased IL-12/23p40 expression, which is opposite to the effect of ERK signaling blockade on IL-12/23p40 in macrophages. This difference in IL-12/23p40 regulation correlated with the differential expression of transcription factors cFos and IRF1, which are known to regulate IL-12 family members, including IL-12 and IL-23. Thus, the impact of ERK signaling in response to TLR2 stimulation differs between macrophages and DCs, potentially regulating their distinctive functions in the immune system. ERK-mediated suppression of IL-12/23p40 in macrophages may prevent excessive inflammation and associated tissue damage following TLR2-stimulation, while ERK-mediated induction of IL-12/23p40 in DCs may promote priming of T helper 1 (Th1) responses. A greater understanding of the role that ERK signaling plays in different immune cell types may inform the development of host-directed therapy and optimal adjuvanticity for a number of infectious pathogens.
Collapse
|
36
|
Ozaki Y, Kishimoto T, Yamashita Y, Kaneko T, Higuchi K, Mae M, Oohira M, Mohammad AI, Yanagiguchi K, Yoshimura A. Expression of osteoclastogenic and anti-osteoclastogenic cytokines differs in mouse gingiva injected with lipopolysaccharide, peptidoglycan, or both. Arch Oral Biol 2020; 122:104990. [PMID: 33259988 DOI: 10.1016/j.archoralbio.2020.104990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Bacterial substances in subgingival biofilm evoke alveolar bone resorption. We previously reported that gingival injection of bacterial lipopolysaccharide (LPS) and peptidoglycan (PGN) induced alveolar bone resorption in mice. However, the mechanism by which LPS and PGN induce osteoclast formation has not been investigated. The aim of this study is to clarify the role of osteoclastogenic and anti-osteoclastogenic cytokines in the alveolar bone resorption induced by LPS and PGN. MATERIALS LPS from Escherichia coli, PGN from Staphylococcus aureus, or both were injected into the gingiva of mice every 48 h for a total of 13 times. Alveolar bone resorption was assessed histochemically by tartrate-resistant acid phosphatase staining. Expression of the receptor activator of nuclear factor-κB ligand (RANKL), tumor necrosis factor (TNF)-α, interleukin (IL)-17, and IL-10 were analyzed by immunostaining. To analyze the role of these cytokines, RANKL-pretreated mouse bone marrow macrophages were stimulated with LPS, PGN, or LPS + PGN with or without anti-TNF-α antibody, IL-17, or IL-10. RESULTS Alveolar bone resorption was induced by both LPS and PGN and exacerbated by LPS + PGN. LPS induced higher RANKL expression than PGN. Expression of TNF-α and IL-10 was correlated with bone resorption. PGN injections induced the strongest expression of IL-17, followed by LPS + PGN and LPS. In an in vitro osteoclastogenesis assay, anti-TNF-α antibody and IL-10 inhibited osteoclast formation, but IL-17 promoted it. CONCLUSION LPS, PGN, or LPS + PGN injections induce distinctive expression of TNF-α, IL-10, and IL-17, suggesting that the composition of these bacterial ligands in dental plaque is critical for alveolar bone resorption.
Collapse
Affiliation(s)
- Yukio Ozaki
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Takaaki Kishimoto
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Kishimoto Dental Office, Oita, Japan
| | - Yasunori Yamashita
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takashi Kaneko
- Center for Oral Diseases, Fukuoka Dental College, Fukuoka, Japan
| | - Kanako Higuchi
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Megumi Mae
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masayuki Oohira
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Alam Ibtehaz Mohammad
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kajiro Yanagiguchi
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Atsutoshi Yoshimura
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
37
|
Kirtland ME, Tsitoura DC, Durham SR, Shamji MH. Toll-Like Receptor Agonists as Adjuvants for Allergen Immunotherapy. Front Immunol 2020; 11:599083. [PMID: 33281825 PMCID: PMC7688745 DOI: 10.3389/fimmu.2020.599083] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/19/2020] [Indexed: 01/19/2023] Open
Abstract
Toll-like receptors (TLRs) are essential components of innate immunity and provide defensive inflammatory responses to invading pathogens. Located within the plasma membranes of cells and also intracellular endosomes, TLRs can detect a range of pathogen associated molecular patterns from bacteria, viruses and fungi. TLR activation on dendritic cells can propagate to an adaptive immune response, making them attractive targets for the development of both prophylactic and therapeutic vaccines. In contrast to conventional adjuvants such as aluminium salts, TLR agonists have a clear immunomodulatory profile that favours anti-allergic T lymphocyte responses. Consequently, the potential use of TLRs as adjuvants in Allergen Immunotherapy (AIT) for allergic rhinitis and asthma remains of great interest. Allergic Rhinitis is a Th2-driven, IgE-mediated disease that occurs in atopic individuals in response to exposure to otherwise harmless aeroallergens such as pollens, house dust mite and animal dander. AIT is indicated in subjects with allergic rhinitis whose symptoms are inadequately controlled by antihistamines and nasal corticosteroids. Unlike anti-allergic drugs, AIT is disease-modifying and may induce long-term disease remission through mechanisms involving upregulation of IgG and IgG4 antibodies, induction of regulatory T and B cells, and immune deviation in favour of Th1 responses that are maintained after treatment discontinuation. This process takes up to three years however, highlighting an unmet need for a more efficacious therapy with faster onset. Agonists targeting different TLRs to treat allergy are at different stages of development. Synthetic TLR4, and TLR9 agonists have progressed to clinical trials, while TLR2, TLR5 and TLR7 agonists been shown to have potent anti-allergic effects in human in vitro experiments and in vivo in animal studies. The anti-allergic properties of TLRs are broadly characterised by a combination of enhanced Th1 deviation, regulatory responses, and induction of blocking antibodies. While promising, a durable effect in larger clinical trials is yet to be observed and further long-term studies and comparative trials with conventional AIT are required before TLR adjuvants can be considered for inclusion in AIT. Here we critically evaluate experimental and clinical studies investigating TLRs and discuss their potential role in the future of AIT.
Collapse
Affiliation(s)
- Max E Kirtland
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma Imperial College London, London, United Kingdom
| | - Daphne C Tsitoura
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen R Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma Imperial College London, London, United Kingdom
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma Imperial College London, London, United Kingdom
| |
Collapse
|
38
|
Effect of Pingchuan Formula on Toll-Like Receptors and Dendritic Cells in an Asthmatic Mouse Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7407016. [PMID: 32953887 PMCID: PMC7481997 DOI: 10.1155/2020/7407016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022]
Abstract
Pingchuan formula (PCF) was created by Professor Yu Jianer. The purpose of this study was to investigate the effect of PCF on dendritic cells (DCs) and toll-like receptors (TLRs) in initiating immunity. A bronchial asthma BALB/c mouse model was established using an OVA excitation method. PCF was immediately administered by gavage after the first excitation. After 7 d, hematoxylin and eosin (HE) staining was used to observe the pathological changes in the asthma model. Eosinophil infiltration and concentrations of IL-4, IFN-r, IL-12, and IFN-α in BALF were determined by enzyme-linked immunosorbent assay (ELISA). Real-time PCR was used to determine mRNA levels of IL-12 and IFN-α. Protein expression levels of ERK, Toll-2, IDO, and Toll-9 were measured by immunoblot. HE and ELISA showed that PCF could improve lung pathological changes and significantly decrease the concentration of IL-4 in BALF. Moreover, PCF could increase IL-12, IFN-α, and IFN-r in BALF. Real-time PCR and western blot showed that PCF restored the DCs and TLRs in initiating immunity. In summary, this study found that PCF can improve the pathological changes and reduce the symptoms of asthma in a BALB/c mouse model. It can facilitate the initiation of immunity by restoring the DCs and TLRs.
Collapse
|
39
|
Voß F, van Beek LF, Schwudke D, Ederveen THA, van Opzeeland FJ, Thalheim D, Werner S, de Jonge MI, Hammerschmidt S. Lipidation of Pneumococcal Antigens Leads to Improved Immunogenicity and Protection. Vaccines (Basel) 2020; 8:vaccines8020310. [PMID: 32560374 PMCID: PMC7350230 DOI: 10.3390/vaccines8020310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/04/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pneumoniae infections lead to high morbidity and mortality rates worldwide. Pneumococcal polysaccharide conjugate vaccines significantly reduce the burden of disease but have a limited range of protection, which encourages the development of a broadly protective protein-based alternative. We and others have shown that immunization with pneumococcal lipoproteins that lack the lipid anchor protects against colonization. Since immunity against S. pneumoniae is mediated through Toll-like receptor 2 signaling induced by lipidated proteins, we investigated the effects of a lipid modification on the induced immune responses in either intranasally or subcutaneously vaccinated mice. Here, we demonstrate that lipidation of recombinant lipoproteins DacB and PnrA strongly improves their immunogenicity. Mice immunized with lipidated proteins showed enhanced antibody concentrations and different induction kinetics. The induced humoral immune response was modulated by lipidation, indicated by increased IgG2/IgG1 subclass ratios related to Th1-type immunity. In a mouse model of colonization, immunization with lipidated antigens led to a moderate but consistent reduction of pneumococcal colonization as compared to the non-lipidated proteins, indicating that protein lipidation can improve the protective capacity of the coupled antigen. Thus, protein lipidation represents a promising approach for the development of a serotype-independent pneumococcal vaccine.
Collapse
Affiliation(s)
- Franziska Voß
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489 Greifswald, Germany; (F.V.); (D.T.); (S.W.)
| | - Lucille F. van Beek
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, 6525 GA Nijmegen, The Netherlands; (L.F.v.B.); (F.J.v.O.); (M.I.d.J.)
- Radboud Center for Infectious Diseases, Radboudumc, 6525 GA Nijmegen, The Netherlands
| | - Dominik Schwudke
- Division of Bioanalytical Chemistry, Priority Area Infection, Research Center Borstel, Leibniz Center for Medicine and Bioscience, 23845 Borstel, Germany;
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
- Airway Research Center North Member of the German Center for Lung Research (DZL), 22927 Großhansdorf, Germany
| | - Thomas H. A. Ederveen
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Fred J. van Opzeeland
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, 6525 GA Nijmegen, The Netherlands; (L.F.v.B.); (F.J.v.O.); (M.I.d.J.)
- Radboud Center for Infectious Diseases, Radboudumc, 6525 GA Nijmegen, The Netherlands
| | - Daniela Thalheim
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489 Greifswald, Germany; (F.V.); (D.T.); (S.W.)
| | - Sidney Werner
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489 Greifswald, Germany; (F.V.); (D.T.); (S.W.)
| | - Marien I. de Jonge
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, 6525 GA Nijmegen, The Netherlands; (L.F.v.B.); (F.J.v.O.); (M.I.d.J.)
- Radboud Center for Infectious Diseases, Radboudumc, 6525 GA Nijmegen, The Netherlands
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489 Greifswald, Germany; (F.V.); (D.T.); (S.W.)
- Correspondence: ; Tel.: +49-383-4420-5700; Fax: +49-3834-4205-709
| |
Collapse
|
40
|
Angeles JMM, Mercado VJP, Rivera PT. Behind Enemy Lines: Immunomodulatory Armamentarium of the Schistosome Parasite. Front Immunol 2020; 11:1018. [PMID: 32582161 PMCID: PMC7295904 DOI: 10.3389/fimmu.2020.01018] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
The deeply rooted, intricate relationship between the Schistosoma parasite and the human host has enabled the parasite to successfully survive within the host and surreptitiously evade the host's immune attacks. The parasite has developed a variety of strategies in its immunomodulatory armamentarium to promote infection without getting harmed or killed in the battlefield of immune responses. These include the production of immunomodulatory molecules, alteration of membranes, and the promotion of granuloma formation. Schistosomiasis thus serves as a paradigm for understanding the Th2 immune responses seen in various helminthiases. This review therefore aims to summarize the immunomodulatory mechanisms of the schistosome parasites to survive inside the host. Understanding these immunomodulatory strategies not only provides information on parasite-host interactions, but also forms the basis in the development of novel drugs and vaccines against the schistosome infection, as well as various types of autoimmune and inflammatory conditions.
Collapse
Affiliation(s)
- Jose Ma M Angeles
- Department of Parasitology, College of Public Health, University of the Philippines Manila, Manila, Philippines
| | - Van Jerwin P Mercado
- Department of Parasitology, College of Public Health, University of the Philippines Manila, Manila, Philippines
| | - Pilarita T Rivera
- Department of Parasitology, College of Public Health, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
41
|
Fang L, Shen Q, Wu H, He F, Ding P, Xu K, Yan X, Wang M, Li S, Liu R. TLR2 favors OVA-induced allergic airway inflammation in mice through JNK signaling pathway with activation of autophagy. Life Sci 2020; 256:117896. [PMID: 32504758 DOI: 10.1016/j.lfs.2020.117896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/21/2020] [Accepted: 05/31/2020] [Indexed: 12/15/2022]
Abstract
AIMS Numerous studies indicate that toll-like receptor 2 (TLR2) led to divergent effects in asthma. The occurrence of autophagy in asthma pathogenesis is still incompletely understood. Here, we aimed to investigate the role of TLR2 and the underlying mechanisms in allergic airway inflammation and autophagy activation. MAIN METHODS C57BL/6 and TLR2 knockout (TLR2-/-) mice were subjected to an ovalbumin (OVA)-immunized allergic airway model, and were treated with SP600125. Differential cell counts in bronchoalveolar lavage fluid were determined by Wright's staining. Histological analysis of airway inflammation was determined by haematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining. The levels of OVA-specific immunoglobulin E (IgE), tumor necrosis factor α (TNF-α) and interleukin 10 (IL-10) were detected by enzyme-linked immunosorbent assay (ELISA). Proteins expression in lung tissues was detected by western blot, expression of TLR2 was further observed by immunofluorescence. Autophagy activation was determined by western blot and transmission electron microscopy (TEM). KEY FINDINGS TLR2 expression was increased upon OVA challenge, and TLR2 deficiency was associated with decreased allergic airway inflammation. Meanwhile, TLR2 deficiency weakened autophagy activation. Moreover, inhibition of c-Jun N-terminal kinase (JNK) by SP600125 also suppressed OVA-induced allergic airway inflammation and autophagy activation. Interestingly, treating TLR2-/- mice with SP600125 showed similar OVA-induced allergic airway inflammation and autophagy activation compared to that in vehicle-treated TLR2-/- mice. SIGNIFICANCE TLR2 might contribute to the maintenance of allergic airway inflammation through JNK signaling pathway accompanying with autophagy activation. These findings may provide a novel signal target for prevention of allergic airway inflammation.
Collapse
Affiliation(s)
- Lei Fang
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Qiying Shen
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Huimei Wu
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Fang He
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; No.1 Department of Respiratory Medicine, Anhui Chest Hospital, Jixi Road 397, Hefei, Anhui 230022, China
| | - Peishan Ding
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Ke Xu
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230031, China
| | - Xuebo Yan
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Muzi Wang
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Shuai Li
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; Intensive Care Unit, The Fourth Affiliated Hospital of Anhui Medical University, Huaihai Avenue 100, Hefei, Anhui 230012, China
| | - Rongyu Liu
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China.
| |
Collapse
|
42
|
Bao L, Hao C, Wang J, Wang D, Zhao Y, Li Y, Yao W. High-Dose Cyclophosphamide Administration Orchestrates Phenotypic and Functional Alterations of Immature Dendritic Cells and Regulates Th Cell Polarization. Front Pharmacol 2020; 11:775. [PMID: 32523537 PMCID: PMC7261842 DOI: 10.3389/fphar.2020.00775] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 12/29/2022] Open
Abstract
High-dose cyclophosphamide (CTX) inhibits the immune response. Dendritic cells (DCs) are professional antigen presenting cells (APCs) with a crucial role in initiating immune responses and sustaining immune tolerance. The relative contribution of DCs to immunosuppression induced by high-dose CTX is not well-documented. In this study, we employed the CTX-induced immunosuppressive rat model to examine alterations in DCs. We generated and cultured monocyte-derived immature DCs (imDCs) in vitro and explored their capacity of antigen uptake, T cell priming, cytokine production, and surface marker expression following high-dose CTX. Subsequently, we co-cultured CTX-treated imDCs with Th cells to determine Th cell polarization, and further explored the Toll-like receptor/Myeloid differentiation primary response 88/Mitogen-activated protein kinase (TLR/MyD88/MAPK) pathway. Our results show reduced cell number and surface maker alterations in splenic CD103+ DCs of CTX-treated immunosuppressed rats. In vitro, high-dose CTX weakened the antigen uptake capacity and enhanced the T cell priming capacity of imDCs, in addition to triggering imDC surface marker alterations. TLR, MyD88, and MAPK expression levels, involved in mediating Th cell polarization, were also significantly elevated. Our collective findings indicate that high-dose CTX administration potentiates phenotypic and functional alterations of imDC. Such changes may contribute to the regulation of Th polarization.
Collapse
Affiliation(s)
- Lei Bao
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, China.,Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China.,Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, China
| | - Changfu Hao
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Juan Wang
- Department of Statistics, Hebei General Hospital, Shijiazhuang, China
| | - Di Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Youliang Zhao
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yiping Li
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wu Yao
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
43
|
Guragain D, Gurung P, Chang JH, Katila N, Chang HW, Jeong BS, Choi DY, Kim JA. AMPK is essential for IL-10 expression and for maintaining balance between inflammatory and cytoprotective signaling. Biochim Biophys Acta Gen Subj 2020; 1864:129631. [PMID: 32418902 DOI: 10.1016/j.bbagen.2020.129631] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) exerts its anti-inflammatory effects by suppressing redox-sensitive nuclear factor kappa B (NF-κB) and pro-inflammatory cytokines including TNF-α. However, it is unclear whether AMPK regulates anti-inflammatory cytokine expressions in the presence of oxidative stress-induced inflammation. We sought to elucidate the mechanisms whereby AMPK regulates inflammatory cytokine expressions under NADPH oxidase (NOX)-induced oxidative stress. METHODS HT-29 human colonic epithelial cells transfected with AMPKα shRNA and mouse models with AMPKα knocked out in epithelial cells (AMPKαfl/fl-Vil-Cre) or macrophages (AMPKαfl/fl-Lyz2-Cre) were used to examine the effects of AMPK and NOX on signaling pathways and cytokine expressions. RESULTS In HT-29 cells, 5-hydroxytryptamine (5-HT)-induced NOX activity was enhanced by AMPKα silencing, and resulted in inflammatory cell death. AMPKα deletion specific for colon epithelial cells (AMPKαfl/fl-Vil-Cre) or macrophages (AMPKαfl/fl-Lyz2-Cre) intensified 5-HT- or dextran sulfate sodium (DSS)-induced upregulations of NOX2, TNF-α, and IL-6, but completely abolished basal and 5-HT- or DSS-induced upregulation of IL-10 in colon epithelium. Furthermore, 5-HT- and DSS-induced changes were accompanied by marked upregulations of increased inflammatory signaling pathways linked to NF-κB, AP-1, and STAT3 transcription factors, and to GATA, a cell fate-directing signaling. In addition, AMPKα deletion significantly fortified 5-HT- or DSS-induced downregulations of cytoprotective signaling pathways (Nrf2, HIF-1α, and KLF4). CONCLUSION Basal AMPKα maintains an anti-inflammatory state by inhibiting NOX, balancing pro-/anti-inflammatory signaling pathways, and directing IL-10 production. When these regulatory roles of AMPK are diminished by oxidative stress, colon epithelium undergoes inflammation despite IL-10 production.
Collapse
Affiliation(s)
- Diwakar Guragain
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Pallavi Gurung
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jae-Hoon Chang
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Nikita Katila
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Hyeun Wook Chang
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Byeong-Seon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
44
|
Koya T, Date I, Kawaguchi H, Watanabe A, Sakamoto T, Togi M, Kato T, Yoshida K, Kojima S, Yanagisawa R, Koido S, Sugiyama H, Shimodaira S. Dendritic Cells Pre-Pulsed with Wilms' Tumor 1 in Optimized Culture for Cancer Vaccination. Pharmaceutics 2020; 12:pharmaceutics12040305. [PMID: 32231023 PMCID: PMC7238244 DOI: 10.3390/pharmaceutics12040305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/21/2022] Open
Abstract
With recent advances in cancer vaccination therapy targeting tumor-associated antigens (TAAs), dendritic cells (DCs) are considered to play a central role as a cell-based drug delivery system in the bioactive immune environment. Ex vivo generation of monocyte-derived DCs has been conventionally applied in adherent manufacturing systems with separate loading of TAAs before clinical use. We developed DCs pre-pulsed with Wilms’ tumor (WT1) peptides in low-adhesion culture maturation (WT1-DCs). Quality tests (viability, phenotype, and functions) of WT1-DCs were performed for process validation, and findings were compared with those for conventional DCs (cDCs). In comparative analyses, WT1-DCs showed an increase in viability and recovery of the DC/monocyte ratio, displaying lower levels of IL-10 (an immune suppressive cytokine) and a similar antigen-presenting ability in an in vitro cytotoxic T lymphocytes (CTLs) assay with cytomegalovirus, despite lower levels of CD80 and PD-L2. A clinical study revealed that WT1-specific CTLs (WT1-CTLs) were detected upon using the WT1-DCs vaccine in patients with cancer. A DC vaccine containing TAAs produced under an optimized manufacturing protocol is a potentially promising cell-based drug delivery system to induce acquired immunity.
Collapse
Affiliation(s)
- Terutsugu Koya
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
- Center for Regenerative medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku 920-0293, Japan;
| | - Ippei Date
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
| | - Haruhiko Kawaguchi
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
| | - Asuka Watanabe
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
| | - Takuya Sakamoto
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
- Center for Regenerative medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku 920-0293, Japan;
| | - Misa Togi
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
- Center for Regenerative medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku 920-0293, Japan;
| | - Tomohisa Kato
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
| | - Kenichi Yoshida
- Center for Regenerative medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku 920-0293, Japan;
| | - Shunsuke Kojima
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan; (S.K.); (R.Y.)
| | - Ryu Yanagisawa
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan; (S.K.); (R.Y.)
| | - Shigeo Koido
- Department of Gastroenterology and Hepatology, The Jikei University School of Medicine, Kashiwa, Chiba 277-8567, Japan;
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan;
| | - Shigetaka Shimodaira
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Kahoku 920-0293, Japan; (T.K.); (I.D.); (H.K.); (A.W.); (T.S.); (M.T.); (T.K.J.)
- Center for Regenerative medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku 920-0293, Japan;
- Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto, Nagano 390-8621, Japan; (S.K.); (R.Y.)
- Correspondence: ; Tel.: +81-76-218-8304
| |
Collapse
|
45
|
Azeem W, Bakke RM, Appel S, Øyan AM, Kalland KH. Dual Pro- and Anti-Inflammatory Features of Monocyte-Derived Dendritic Cells. Front Immunol 2020; 11:438. [PMID: 32292402 PMCID: PMC7120039 DOI: 10.3389/fimmu.2020.00438] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/25/2020] [Indexed: 01/01/2023] Open
Abstract
The transcription factor β-catenin is able to induce tolerogenic/anti-inflammatory features in different types of dendritic cells (DCs). Monocyte-derived dendritic cells (moDCs) have been widely used in dendritic cell-based cancer therapy, but so far with limited clinical efficacy. We wanted to investigate the hypothesis that aberrant differentiation or induction of dual pro- and anti-inflammatory features may be β-catenin dependent in moDCs. β-catenin was detectable in both immature and lipopolysaccharide (LPS)-stimulated DCs. The β-catenin inhibitor ICG-001 dose-dependently increased the pro-inflammatory signature cytokine IL-12p70 and decreased the anti-inflammatory signature molecule IL-10. The β-catenin activator 6-bromoindirubin-3′-oxime (6-BIO) dose-dependently increased total and nuclear β-catenin, and this was associated with decreased IL-12p70, increased IL-10, and reduced surface expression of activation markers, such as CD80 and CD86, and increased expression of inhibitory markers, such as PD-L1. 6-BIO and ICG-001 competed dose-dependently regarding these features. Genome-wide mRNA expression analyses further underscored the dual development of pro- and anti-inflammatory features of LPS-matured moDCs and suggest a role for β-catenin inhibition in production of more potent therapeutic moDCs.
Collapse
Affiliation(s)
- Waqas Azeem
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ragnhild Maukon Bakke
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway
| | - Silke Appel
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Broegelmann Research Laboratory, University of Bergen, Bergen, Norway
| | - Anne Margrete Øyan
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Helse Bergen, Bergen, Norway
| | - Karl-Henning Kalland
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway.,Norway Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| |
Collapse
|
46
|
Arora P, Moll JM, Andersen D, Workman CT, Williams AR, Kristiansen K, Brix S. Body fluid from the parasitic worm Ascaris suum inhibits broad-acting pro-inflammatory programs in dendritic cells. Immunology 2020; 159:322-334. [PMID: 31705653 PMCID: PMC7011627 DOI: 10.1111/imm.13151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) are essential for generating T-cell-based immune responses through sensing of potential inflammatory and metabolic cues in the local environment. However, there is still limited insight into the processes defining the resultant DC phenotype, including the type of early transcriptional changes in pro-inflammatory cues towards regulatory or type 2 immune-based cues induced by a variety of exogenous and endogenous molecules. Here we compared the ability of a selected number of molecules to modulate the pro-inflammatory phenotype of lipopolysaccharide (LPS) and interferon-γ (IFN-γ)-stimulated human monocyte-derived DCs towards an anti-inflammatory or regulatory phenotype, including Ascaris suum body fluid [helminth pseudocoelomic fluid (PCF)], the metabolites succinate and butyrate, and the type 2 cytokines thymic stromal lymphopoietin and interleukin-25. Our data show that helminth PCF and butyrate treatment suppress the T helper type 1 (Th1)-inducing pro-inflammatory DC phenotype through induction of different transcriptional programs in DCs. RNA sequencing indicated that helminth PCF treatment strongly inhibited the Th1 and Th17 polarizing ability of LPS + IFN-γ-matured DCs by down-regulating myeloid differentiation primary response gene 88 (MyD88)-dependent and MyD88-independent pathways in Toll-like receptor 4 signaling. By contrast, butyrate treatment had a strong Th1-inhibiting action, and transcripts encoding important gut barrier defending factors such as IL18, IL1B and CXCL8 were up-regulated. Collectively, our results further understanding of how compounds from parasites and gut microbiota-derived butyrate may exert immunomodulatory effects on the host immune system.
Collapse
Affiliation(s)
- Pankaj Arora
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Janne Marie Moll
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Daniel Andersen
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | | | - Andrew R. Williams
- Parasitology and Aquatic PathobiologyUniversity of CopenhagenCopenhagenDenmark
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular BiomedicineDepartment of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Susanne Brix
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| |
Collapse
|
47
|
Wang M, Luo Y, Sun T, Mao C, Jiang Y, Yu X, Li Z, Xie T, Wu F, Yan H, Teng L. The Ectopic Expression of SurvivinT34A and FilC Can Enhance the Oncolytic Effects of Vaccinia Virus in Murine Gastric Cancer. Onco Targets Ther 2020; 13:1011-1025. [PMID: 32099404 PMCID: PMC7006861 DOI: 10.2147/ott.s230902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/16/2020] [Indexed: 01/13/2023] Open
Abstract
Background/Aims Anti-tumor vaccines have been shown to be effective in cancer therapeutics ever since the anti-HPV vaccine was developed. Compared to conventional chemotherapy, anti-tumor vaccines can specifically target cancer cells and they have lower side effects. We developed a recombinant vaccinia virus (VACV) (Western Reserve) WR strain, and we tested its anti-tumor effects in an animal model. Methods A recombinant VACV WR strain expressing mutant survivin T34A (SurT34A) and FilC was constructed and validated. Its oncolytic effect was tested in vitro using a CCK-8 assay, and its tolerance and anti-tumor effects were tested in a murine gastric cancer model. The proportion of lymphocytes in the spleen and tumor was determined after antibody-mediated immuno-depletion. Results The recombinant VACV showed a stronger replication ability in tumor cells, and it was safe in vivo, even at high doses. The combination of vv-SurT34A and vv-FilC resulted in a stronger anti-tumor effect compared to either construct alone. However, the inhibitory effect of vv-SurT34A was stronger than the combination. The recombinant VACV activated the host immune response, as indicated by lymphocyte infiltration in the spleen and tumor tissues. Conclusion The recombinant VACV WR strain expressing SurT34A and FilC is a safe and effective anti-tumor vaccine.
Collapse
Affiliation(s)
- Minglong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yanxi Luo
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, People's Republic of China
| | - Ting Sun
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, People's Republic of China
| | - Chenyu Mao
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yili Jiang
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, People's Republic of China
| | - Xiongfei Yu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhongqi Li
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Tian Xie
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Fusheng Wu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Hui Yan
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, People's Republic of China.,Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
48
|
Diaz-Dinamarca DA, Manzo RA, Soto DA, Avendaño-Valenzuela MJ, Bastias DN, Soto PI, Escobar DF, Vasquez-Saez V, Carrión F, Pizarro-Ortega MS, Wilson CAM, Berrios J, Kalergis AM, Vasquez AE. Surface Immunogenic Protein of Streptococcus Group B is an Agonist of Toll-Like Receptors 2 and 4 and a Potential Immune Adjuvant. Vaccines (Basel) 2020; 8:vaccines8010029. [PMID: 31963234 PMCID: PMC7157747 DOI: 10.3390/vaccines8010029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/20/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022] Open
Abstract
Vaccine-induced protection against pathogens, especially subunit-based vaccines, are related to antigen properties but mainly in their ability to stimulate the immune system by the use of an adjuvant. Modern vaccines are formulated with a high level of antigen purity, where an efficient adjuvant is necessary. In this context, the use of protein Toll-Like Receptor (TLR) agonists as vaccine adjuvants has been highlighted because of their optimal immunogenicity and minimal toxicity. The Surface Immunogenic Protein (SIP) from Group B Streptococcus (GBS) has gained importance as a new potential protein-based vaccine. Recently, we reported that recombinant SIP (rSIP) expressed by E. coli and purified by High Performance Liquid Chromatography (HPLC) alone induces a protective humoral immune response. In this study, we present the immunomodulatory properties of rSIP as a protein-based adjuvant, as an agonist of TLR. To this end, we showed that C57BL/6 bone marrow-derived dendritic cells pulsed by rSIP resulted in enhanced CD40, CD80, CD86, and Major Histocompatibility Complex (MHC) class II as well as increased secretion proinflammatory cytokines Interleukin (IL)-6, Interferon (IFN)-γ, Tumor Necrosis Factor (TNF)-α, and IL-10. Next, we investigated the in vivo effect of rSIP in the absence or presence of ovalbumin (OVA) on antigen-specific antibody secretion in C57BL/6 mice. Immunization with rSIP plus OVA showed that anti-OVA IgG2a and IgG1a increased significantly compared with OVA alone in C57BL/6 mice. Also, the immunization of rSIP plus OVA generates increased serum cytokines levels characterized by IL-12p70, IL-10, IL-4, and IFN-γ. Interestingly, we observed that rSIP stimulate Toll Like Receptor (TLR)2 and TLR4, individually expressed by Human embryonic kidney (HEK) 293-derived TLR reporter cells. These findings suggest that rSIP is a new potential protein TLR agonist adjuvant and may be employed in the development of new vaccines.
Collapse
Affiliation(s)
- Diego A. Diaz-Dinamarca
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
| | - Ricardo A. Manzo
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Daniel A. Soto
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - María José Avendaño-Valenzuela
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
| | - Diego N. Bastias
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomas, Santiago 8320000, Chile
| | - Paulina I. Soto
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Daniel F. Escobar
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Valeria Vasquez-Saez
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Flavio Carrión
- Programa de Inmunología Traslacional, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Magdalena S. Pizarro-Ortega
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
| | - Christian A. M. Wilson
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8320000, Chile;
| | - Julio Berrios
- Escuela de Ingeniería en Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile;
| | - Alexis M. Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Abel E. Vasquez
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomas, Santiago 8320000, Chile
- Facultad de Ciencia, Universidad San Sebastián, Providencia, Santiago 8320000, Chile
- Correspondence: ; Tel.: +56-2-2575-5513
| |
Collapse
|
49
|
Aalberse RC, Grüber C, Ljungman M, Kakat S, Wahn U, Niggemann B, Nilsson LJ. Further investigations of the IgE response to tetanus and diphtheria following covaccination with acellular rather than cellular Bordetella pertussis. Pediatr Allergy Immunol 2019; 30:841-847. [PMID: 31419322 DOI: 10.1111/pai.13113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND It has previously been shown in an uncontrolled study that the IgE response to vaccine antigens is downregulated by co-vaccination with cellular Bordetella pertussis vaccine. METHODS In the present study, we compared in a controlled trial the humoral immune response to diphtheria toxoid (D) and tetanus toxoid (T) in relation to co-vaccinated cellular or acellular B pertussis vaccine. IgE, IgG4, and IgG to D and T were analyzed at 2, 7, and 12 months of age in sera of children vaccinated with D and T (DT, N = 68), cellular (DTPw, N = 68), 2- or 5-component acellular B pertussis vaccine (DTPa2, N = 64; DTPa5, N = 65). RESULTS One month after vaccination, D-IgE was detected in 10% sera of DTPw-vaccinated children, whereas vaccination in the absence of whole-cell pertussis resulted in 50%-60% IgE positivity. Six months after vaccination, the IgE antibody levels were found to be more persistent than the IgG antibodies. These diphtheria findings were mirrored by those for tetanus. Only minor differences between vaccine groups were found with regard to D-IgG and T-IgG. No immediate-type allergic reactions were observed. CONCLUSION Cellular (but not acellular) B pertussis vaccine downregulates IgE to co-vaccinated antigens in infants. We assume that the absence of immediate-type allergic reactions is due to the high levels of IgG antibodies competing with IgE antibodies.
Collapse
Affiliation(s)
- Rob C Aalberse
- Amsterdam and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Christoph Grüber
- Department of Pediatric Pneumology and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Suzan Kakat
- Department of Pediatric Pneumology and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrich Wahn
- Department of Pediatric Pneumology and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bodo Niggemann
- Department of Pediatric Pneumology and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lennart J Nilsson
- Department of Clinical and Experimental Medicine, Allergy Center, Linköping University, Linköping, Sweden
| |
Collapse
|
50
|
Jehangir A, Shahzad M, Shahid K, Waheed A, Ayub F. Zinc and iron complexes of oleanolic acid, (OA) attenuate allergic airway inflammation in rats. Inflammopharmacology 2019; 27:1179-1192. [PMID: 31069605 DOI: 10.1007/s10787-019-00597-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022]
Abstract
Oleanolic acid (OA) is a hydroxyl pentacyclic triterpene acid (HTAs) used in various ailments. Inflammatory diseases may be profoundly influenced by iron (Fe) and zinc (Zn) status. We studied the anti-asthmatic effects of two metal complexes (Fe and Zn) of OA in the ovalbumin (OVA)-induced rat model. Delayed type hypersensitivity (DTH) was measured. Total and differential leucocyte count was done in blood as well as bronchoalveolar lavage fluid (BALF). The mRNA expression levels of pro-inflammatory cytokines were measured in lung tissue by reverse transcription polymerase chain reaction. The levels of cyclooxygenase-2 (COX-2), immunoglobulin E (IgE) and 5-lipoxygenase (5-LOX) were estimated by enzyme linked immunosorbent assay. Splenocyte proliferation was performed through BrdU uptake method and nitric oxide levels were measured by colorimetric assay kit. The acute toxicity study was also done for the complexes. The asthmatic group developed allergic airway inflammation shown by increased DTH and inflammatory markers in blood and BALF. OA + Fe and OA + Zn displayed significant decrease in DTH, NO, expression of IL-4, 5, 13, 17, toll-like receptor-2, nuclear factor-kappa B and tumor necrosis factor-α; serum IgE, COX-2, and 5-LOX. The metal complexes also attenuated OVA-stimulated splenocyte proliferation. While no hepatotoxic or nephrotoxic potential was shown by OA + Fe and OA + Zn. Our findings indicate that both OA + Fe and OA + Zn possess significant anti-asthmatic effect which may be ascribed to its immunomodulatory and anti-inflammatory features.
Collapse
Affiliation(s)
- Adnan Jehangir
- Department of Pharmacology, University of Health Sciences, Lahore, 54600, Pakistan
- Riphah International University, Islamabad, Pakistan
| | - Muhammad Shahzad
- Department of Pharmacology, University of Health Sciences, Lahore, 54600, Pakistan.
| | | | - Akbar Waheed
- Riphah International University, Islamabad, Pakistan
| | - Farhana Ayub
- Riphah International University, Islamabad, Pakistan
| |
Collapse
|