1
|
Wu X, Zhang J, Deng Z, Sun X, Zhang Y, Zhang C, Wang J, Yu X, Yang G. Bacteria-based biohybrids for remodeling adenosine-mediated immunosuppression to boost radiotherapy-triggered antitumor immune response. Biomaterials 2025; 316:123000. [PMID: 39674101 DOI: 10.1016/j.biomaterials.2024.123000] [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: 07/17/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Radiotherapy (RT) can trigger immunogenic cell death (ICD) in tumor cells and release adenosine triphosphate (ATP) to activate antitumor immunity. However, the formation of immunosuppressive adenosine (ADO) mediated by ectonucleotidases including CD39 and CD73, can exacerbate the immunosuppressive effects. Herein, a radiosensitizer-based metal-organic framework (MOF) composed of bismuth (Bi) and ellagic acid (EA) was synthesized in situ on the surface of Escherichia coli Nissle 1917 (EcN) to serve as a carrier for the CD39 inhibitor sodium polyoxotungstate (POM-1). This therapeutic platform, acting as a radiosensitizer, significantly enhances cytotoxicity against tumor cells while effectively inducing ICD and releasing high concentrations of ATP. Subsequently, the released POM-1 increases the levels of pro-inflammatory extracellular ATP while preventing tumor immunosuppression caused by the accumulation of ADO. Additionally, as a natural immune adjuvant, EcN further promotes the maturation of dendritic cells (DCs) and the infiltration of cytotoxic T lymphocytes (CTLs). As a result, such treatment initiates the destruction of established tumor growth and induces strong abscopal effects, leading to a significant inhibition of tumor metastases. This strategy presents a bacterial-based biohybrid system that facilitates RT-induced ICD while simultaneously limiting the degradation of ATP into ADO, thereby achieving sustained anti-tumor immunity.
Collapse
Affiliation(s)
- Xirui Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou 215004, China
| | - Zheng Deng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xianglong Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yifan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiadong Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xinke Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
2
|
Kumar A, Dixit S, Srinivasan K, M D, Vincent PMDR. Personalized cancer vaccine design using AI-powered technologies. Front Immunol 2024; 15:1357217. [PMID: 39582860 PMCID: PMC11581883 DOI: 10.3389/fimmu.2024.1357217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 09/24/2024] [Indexed: 11/26/2024] Open
Abstract
Immunotherapy has ushered in a new era of cancer treatment, yet cancer remains a leading cause of global mortality. Among various therapeutic strategies, cancer vaccines have shown promise by activating the immune system to specifically target cancer cells. While current cancer vaccines are primarily prophylactic, advancements in targeting tumor-associated antigens (TAAs) and neoantigens have paved the way for therapeutic vaccines. The integration of artificial intelligence (AI) into cancer vaccine development is revolutionizing the field by enhancing various aspect of design and delivery. This review explores how AI facilitates precise epitope design, optimizes mRNA and DNA vaccine instructions, and enables personalized vaccine strategies by predicting patient responses. By utilizing AI technologies, researchers can navigate complex biological datasets and uncover novel therapeutic targets, thereby improving the precision and efficacy of cancer vaccines. Despite the promise of AI-powered cancer vaccines, significant challenges remain, such as tumor heterogeneity and genetic variability, which can limit the effectiveness of neoantigen prediction. Moreover, ethical and regulatory concerns surrounding data privacy and algorithmic bias must be addressed to ensure responsible AI deployment. The future of cancer vaccine development lies in the seamless integration of AI to create personalized immunotherapies that offer targeted and effective cancer treatments. This review underscores the importance of interdisciplinary collaboration and innovation in overcoming these challenges and advancing cancer vaccine development.
Collapse
Affiliation(s)
- Anant Kumar
- School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Shriniket Dixit
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Kathiravan Srinivasan
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Dinakaran M
- School of Computer Science and Engineering, Vellore Institute of Technology, Chennai, India
| | - P. M. Durai Raj Vincent
- School of Computer Science Engineering and Information Systems, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
3
|
Ma L, Yang H, Wu S, Wang C, Mei J. DPP7 as a Potential Therapeutic Marker for Colorectal Cancer. J Cancer 2024; 15:5425-5439. [PMID: 39247602 PMCID: PMC11375546 DOI: 10.7150/jca.93112] [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: 12/11/2023] [Accepted: 07/09/2024] [Indexed: 09/10/2024] Open
Abstract
Background: Dipeptidyl peptidase 7 (DPP7) is overexpressed in various tumors, but its role in colorectal cancer (CRC) remains unclear. Study the Impact of DPP7 on malignant progression and tumor immunity in CRC. Methods: We utilized Tumor Immune Estimation Resource 2.0 (TIMER2.0) and The Cancer Genome Atlas (TCGA) analyses to assess the expression of DPP7 in tumors and validated it through immunohistochemistry and immunoblotting. Additionally, we investigated the relationship between DPP7 and immune cell infiltration using single-sample Gene Set Enrichment Analysis (ssGSEA) analysis. Finally, the impact of DPP7 on cell proliferation, invasion, migration, and immune cell function in the tumor microenvironment was confirmed through cell experiments and animal studies. Results: DPP7 is highly expressed in CRC, and high expression of DPP7 is associated with poor prognosis. Cell experiments demonstrate that overexpression of DPP7 enhances the proliferation, migration, and invasion capabilities of colorectal cancer cells both in vitro and in vivo. Immune infiltration analysis and co-culture results indicate that overexpression of DPP7 suppresses the immune cell's cytotoxic function against tumors in the tumor microenvironment. Conclusions: DPP7 promotes the malignant potential of colorectal cancer cells and inhibits tumor immune function, thereby promoting the progression of colorectal cancer.
Collapse
Affiliation(s)
- Li Ma
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Molecular Pathology, Nanchang University, Nanchang, China
| | - Hailang Yang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuwei Wu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chunliang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jinhong Mei
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Molecular Pathology, Nanchang University, Nanchang, China
| |
Collapse
|
4
|
He S, Gubin MM, Rafei H, Basar R, Dede M, Jiang X, Liang Q, Tan Y, Kim K, Gillison ML, Rezvani K, Peng W, Haymaker C, Hernandez S, Solis LM, Mohanty V, Chen K. Elucidating immune-related gene transcriptional programs via factorization of large-scale RNA-profiles. iScience 2024; 27:110096. [PMID: 38957791 PMCID: PMC11217617 DOI: 10.1016/j.isci.2024.110096] [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: 12/14/2023] [Revised: 04/03/2024] [Accepted: 05/21/2024] [Indexed: 07/04/2024] Open
Abstract
Recent developments in immunotherapy, including immune checkpoint blockade (ICB) and adoptive cell therapy (ACT), have encountered challenges such as immune-related adverse events and resistance, especially in solid tumors. To advance the field, a deeper understanding of the molecular mechanisms behind treatment responses and resistance is essential. However, the lack of functionally characterized immune-related gene sets has limited data-driven immunological research. To address this gap, we adopted non-negative matrix factorization on 83 human bulk RNA sequencing (RNA-seq) datasets and constructed 28 immune-specific gene sets. After rigorous immunologist-led manual annotations and orthogonal validations across immunological contexts and functional omics data, we demonstrated that these gene sets can be applied to refine pan-cancer immune subtypes, improve ICB response prediction and functionally annotate spatial transcriptomic data. These functional gene sets, informing diverse immune states, will advance our understanding of immunology and cancer research.
Collapse
Affiliation(s)
- Shan He
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew M. Gubin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Merve Dede
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xianli Jiang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qingnan Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yukun Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kunhee Kim
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maura L. Gillison
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiyi Peng
- Department of Biology and Biochemistry, The University of Houston, Houston, TX, USA
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sharia Hernandez
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa M. Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
He S, Gubin MM, Rafei H, Basar R, Dede M, Jiang X, Liang Q, Tan Y, Kim K, Gillison ML, Rezvani K, Peng W, Haymaker C, Hernandez S, Solis LM, Mohanty V, Chen K. Elucidating immune-related gene transcriptional programs via factorization of large-scale RNA-profiles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593433. [PMID: 38798470 PMCID: PMC11118452 DOI: 10.1101/2024.05.10.593433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Recent developments in immunotherapy, including immune checkpoint blockade (ICB) and adoptive cell therapy, have encountered challenges such as immune-related adverse events and resistance, especially in solid tumors. To advance the field, a deeper understanding of the molecular mechanisms behind treatment responses and resistance is essential. However, the lack of functionally characterized immune-related gene sets has limited data-driven immunological research. To address this gap, we adopted non-negative matrix factorization on 83 human bulk RNA-seq datasets and constructed 28 immune-specific gene sets. After rigorous immunologist-led manual annotations and orthogonal validations across immunological contexts and functional omics data, we demonstrated that these gene sets can be applied to refine pan-cancer immune subtypes, improve ICB response prediction and functionally annotate spatial transcriptomic data. These functional gene sets, informing diverse immune states, will advance our understanding of immunology and cancer research.
Collapse
|
6
|
Zahedipour F, Jamialahmadi K, Zamani P, Reza Jaafari M. Improving the efficacy of peptide vaccines in cancer immunotherapy. Int Immunopharmacol 2023; 123:110721. [PMID: 37543011 DOI: 10.1016/j.intimp.2023.110721] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Peptide vaccines have shown great potential in cancer immunotherapy by targeting tumor antigens and activating the patient's immune system to mount a specific response against cancer cells. However, the efficacy of peptide vaccines in inducing a sustained immune response and achieving clinical benefit remains a major challenge. In this review, we discuss the current status of peptide vaccines in cancer immunotherapy and strategies to improve their efficacy. We summarize the recent advancements in the development of peptide vaccines in pre-clinical and clinical settings, including the use of novel adjuvants, neoantigens, nano-delivery systems, and combination therapies. We also highlight the importance of personalized cancer vaccines, which consider the unique genetic and immunological profiles of individual patients. We also discuss the strategies to enhance the immunogenicity of peptide vaccines such as multivalent peptides, conjugated peptides, fusion proteins, and self-assembled peptides. Although, peptide vaccines alone are weak immunogens, combining peptide vaccines with other immunotherapeutic approaches and developing novel approaches such as personalized vaccines can be promising methods to significantly enhance their efficacy and improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Boyle T, O'Lone E, Phua E, Anderson J, Mather A, Fernando SL. Subsequent COVID-19 Prophylaxis in COVID-19 Associated Glomerulopathies. Vaccines (Basel) 2023; 11:1152. [PMID: 37514968 PMCID: PMC10385225 DOI: 10.3390/vaccines11071152] [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: 05/12/2023] [Revised: 06/16/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Successful vaccination has been the decisive factor in the overall decline of SARS-CoV2 infection related morbidity and mortality. However, global effects of the COVID-19 pandemic are ongoing, with reports of glomerular disease occurring in relation to both infection and vaccination. A particular rise in anti-GBM disease has been identified. Information is still emerging regarding the optimal management of such cases. We reviewed anti-GBM antibody detection rates at our test center over the past 5 years. We followed three patients with biopsy confirmed glomerular disease temporally related to COVID-19 vaccination. Each patient proceeded to receive subsequent COVID-19 vaccination as per immunologist recommendations. Further assessment included COVID-19 antibody testing in each case. A three-fold increase in significant anti-GBM antibody results noted at our center was associated with COVID infection in 10% of cases, and COVID vaccination in 25% of cases. We demonstrated that subsequent vaccination did not appear to lead to adverse effects including relapse in our three cases of COVID-19 vaccine-associated GN. We also identified positive COVID-19 antibody levels in two out of three cases, despite immunosuppression. We report a rise in anti-GBM antibody disease incidence. Our small study suggests that COVID-19 antibody testing can help determine COVID prophylaxis requirements, and subsequent vaccination with an alternative vaccine type appears safe.
Collapse
Affiliation(s)
- Therese Boyle
- Department of Clinical Immunology and Allergy, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia
| | - Emma O'Lone
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia
- Department of Renal Medicine, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
| | - Elaine Phua
- Department of Renal Medicine, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
| | - Janet Anderson
- Immunology Laboratory, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
| | - Amanda Mather
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia
- Department of Renal Medicine, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
| | - Suran L Fernando
- Department of Clinical Immunology and Allergy, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia
- Immunology Laboratory, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
| |
Collapse
|
8
|
Jonny J, Putranto TA, Sitepu EC, Irfon R. Dendritic cell vaccine as a potential strategy to end the COVID-19 pandemic. Why should it be Ex Vivo? Expert Rev Vaccines 2022; 21:1111-1120. [PMID: 35593184 DOI: 10.1080/14760584.2022.2080658] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Developing a safe and efficacious vaccine that can induce broad and long-term immunity for SARS-CoV-2 infection is the most critical research to date. As the most potent APCs, dendritic cells (DCs) can induce a robust T cell immunity. In addition, DCs also play an essential role in COVID-19 pathogenesis, making them a potential vaccination target. However, the DCs-based vaccine with ex vivo loading has not yet been explored for COVID-19. AREAS COVERED This review aims to provide the rationale for developing a DCs-based vaccine with ex vivo loading of SARS-CoV-2 antigen. Here, we discuss the role of DCs in immunity and the effect of SARS-CoV-2 infection on DCs. Then, we propose the mechanism of the DCs-based vaccine in inducing immunity and highlight the benefits of ex vivo loading of antigen. EXPERT OPINION We make the case that an ex vivo loaded DC-based vaccination is appropriate for COVID-19 prevention.
Collapse
Affiliation(s)
- Jonny Jonny
- Cellcure Center, Gatot Soebroto Central Army Hospital, Jakarta, Indonesia
| | | | | | - Raoulian Irfon
- Cellcure Center, Gatot Soebroto Central Army Hospital, Jakarta, Indonesia
| |
Collapse
|
9
|
Zhang J, Li Z, Chandrasekar A, Li S, Ludolph A, Boeckers TM, Huber-Lang M, Roselli F, Olde Heuvel F. Fast Maturation of Splenic Dendritic Cells Upon TBI Is Associated With FLT3/FLT3L Signaling. Front Immunol 2022; 13:824459. [PMID: 35281004 PMCID: PMC8907149 DOI: 10.3389/fimmu.2022.824459] [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: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
The consequences of systemic inflammation are a significant burden after traumatic brain injury (TBI), with almost all organs affected. This response consists of inflammation and concurrent immunosuppression after injury. One of the main immune regulatory organs, the spleen, is highly interactive with the brain. Along this brain–spleen axis, both nerve fibers as well as brain-derived circulating mediators have been shown to interact directly with splenic immune cells. One of the most significant comorbidities in TBI is acute ethanol intoxication (EI), with almost 40% of patients showing a positive blood alcohol level (BAL) upon injury. EI by itself has been shown to reduce proinflammatory mediators dose-dependently and enhance anti-inflammatory mediators in the spleen. However, how the splenic immune modulatory effect reacts to EI in TBI remains unclear. Therefore, we investigated early splenic immune responses after TBI with and without EI, using gene expression screening of cytokines and chemokines and fluorescence staining of thin spleen sections to investigate cellular mechanisms in immune cells. We found a strong FLT3/FLT3L induction 3 h after TBI, which was enhanced by EI. The FLT3L induction resulted in phosphorylation of FLT3 in CD11c+ dendritic cells, which enhanced protein synthesis, maturation process, and the immunity of dendritic cells, shown by pS6, peIF2A, MHC-II, LAMP1, and CD68 by immunostaining and TNF-α expression by in-situ hybridization. In conclusion, these data indicate that TBI induces a fast maturation and immunity of dendritic cells which is associated with FLT3/FLT3L signaling and which is enhanced by EI prior to TBI.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| | - Zhenghui Li
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany.,Department of Neurosurgery, Kaifeng Central Hospital, Kaifeng, China
| | - Akila Chandrasekar
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| | - Shun Li
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE) , Ulm, Germany
| | - Tobias Maria Boeckers
- German Center for Neurodegenerative Diseases (DZNE) , Ulm, Germany.,Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital, Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE) , Ulm, Germany.,Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Florian Olde Heuvel
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| |
Collapse
|
10
|
Losada Méndez J, Palomares F, Gómez F, Ramírez-López P, Ramos-Soriano J, Torres MJ, Mayorga C, Rojo J. Immunomodulatory Response of Toll-like Receptor Ligand-Peptide Conjugates in Food Allergy. ACS Chem Biol 2021; 16:2651-2664. [PMID: 34761908 PMCID: PMC8609526 DOI: 10.1021/acschembio.1c00765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Covalent conjugation
of allergens to toll-like receptor (TLR) agonists
appears to be a powerful strategy for the development of safety compounds
for allergen-specific immunomodulatory response toward tolerance in
allergy. In this work, we have synthesized two family of ligands,
an 8-oxoadenine derivative as a ligand for TLR7 and a pyrimido[5,4-b]indole as a ligand for TLR4, both conjugated with a T-cell
peptide of Pru p 3 allergen, the lipid transfer protein (LTP) responsible
for LTP-dependent food allergy. These conjugates interact with dendritic
cells, inducing their specific maturation, T-cell proliferation, and
cytokine production in peach allergic patients. Moreover, they increased
the Treg-cell frequencies in these patients and could induce the IL-10
production. These outcomes were remarkable in the case of the TLR7
ligand conjugated with Pru p 3, opening the door for the potential
application of these allergen–adjuvant systems in food allergy
immunotherapy.
Collapse
Affiliation(s)
- Jorge Losada Méndez
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC─Universidad de Sevilla, 41092 Seville, Spain
| | - Francisca Palomares
- Allergy Unit, IBIMA, Regional University Hospital of Malaga, UMA, 29009 Malaga, Spain
| | - Francisca Gómez
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
| | - Pedro Ramírez-López
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC─Universidad de Sevilla, 41092 Seville, Spain
| | - Javier Ramos-Soriano
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC─Universidad de Sevilla, 41092 Seville, Spain
| | - Maria Jose Torres
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
- Nanostructures for Diagnosing and Treatment of Allergic Diseases Laboratory, Centro Andaluz de Nanomedicina y Biotecnología-BIONAND, 29590 Málaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain
- Medicine Department, Universidad de Málaga-UMA, 29009 Málaga, Spain
| | - Cristobalina Mayorga
- Allergy Unit, IBIMA, Regional University Hospital of Malaga, UMA, 29009 Malaga, Spain
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
- Nanostructures for Diagnosing and Treatment of Allergic Diseases Laboratory, Centro Andaluz de Nanomedicina y Biotecnología-BIONAND, 29590 Málaga, Spain
| | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC─Universidad de Sevilla, 41092 Seville, Spain
| |
Collapse
|
11
|
Zhuang B, Shang J, Yao Y. HLA-G: An Important Mediator of Maternal-Fetal Immune-Tolerance. Front Immunol 2021; 12:744324. [PMID: 34777357 PMCID: PMC8586502 DOI: 10.3389/fimmu.2021.744324] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/11/2021] [Indexed: 01/17/2023] Open
Abstract
Maternal-fetal immune-tolerance occurs throughout the whole gestational trimester, thus a mother can accept a genetically distinct fetus without immunological aggressive behavior. HLA-G, one of the non-classical HLA class I molecules, is restricted-expression at extravillous trophoblast. It can concordantly interact with various kinds of receptors mounted on maternally immune cells residing in the uterus (e.g. CD4+ T cells, CD8+ T cells, natural killer cells, macrophages, and dendritic cells) for maintaining immune homeostasis of the maternal-fetus interface. HLA-G is widely regarded as the pivotal protective factor for successful pregnancies. In the past 20 years, researches associated with HLA-G have been continually published. Indeed, HLA-G plays a mysterious role in the mechanism of maternal-fetal immune-tolerance. It can also be ectopically expressed on tumor cells, infected sites and other pathologic microenvironments to confer a significant local tolerance. Understanding the characteristics of HLA-G in immunologic tolerance is not only beneficial for pathological pregnancy, but also helpful to the therapy of other immune-related diseases, such as organ transplant rejection, tumor migration, and autoimmune disease. In this review, we describe the biological properties of HLA-G, then summarize our understanding of the mechanisms of fetomaternal immunologic tolerance and the difference from transplant tolerance. Furthermore, we will discuss how HLA-G contributes to the tolerogenic microenvironment during pregnancy. Finally, we hope to find some new aspects of HLA-G in fundamental research or clinical application for the future.
Collapse
Affiliation(s)
- Baimei Zhuang
- Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China.,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jin Shang
- Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuanqing Yao
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynecology, The First Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
12
|
Stephens AJ, Burgess-Brown NA, Jiang S. Beyond Just Peptide Antigens: The Complex World of Peptide-Based Cancer Vaccines. Front Immunol 2021; 12:696791. [PMID: 34276688 PMCID: PMC8279810 DOI: 10.3389/fimmu.2021.696791] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022] Open
Abstract
Peptide-based cancer vaccines rely upon the strong activation of the adaptive immune response to elicit its effector function. They have shown to be highly specific and safe, but have yet to prove themselves as an efficacious treatment for cancer in the clinic. This is for a variety of reasons, including tumour heterogeneity, self-tolerance, and immune suppression. Importance has been placed on the overall design of peptide-based cancer vaccines, which have evolved from simple peptide derivatives of a cancer antigen, to complex drugs; incorporating overlapping regions, conjugates, and delivery systems to target and stimulate different components of antigen presenting cells, and to bolster antigen cross-presentation. Peptide-based cancer vaccines are increasingly becoming more personalised to an individual's tumour antigen repertoire and are often combined with existing cancer treatments. This strategy ultimately aids in combating the shortcomings of a more generalised vaccine strategy and provides a comprehensive treatment, taking into consideration cancer cell variability and its ability to avoid immune interrogation.
Collapse
Affiliation(s)
- Alexander J Stephens
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Nicola A Burgess-Brown
- Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Shisong Jiang
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Yan Y, Yao D, Li X. Immunological Mechanism and Clinical Application of PAMP Adjuvants. Recent Pat Anticancer Drug Discov 2021; 16:30-43. [PMID: 33563182 DOI: 10.2174/1574892816666210201114712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The host innate immune system can recognize Pathogen-Associated Molecular Patterns (PAMPs) through Pattern Recognition Receptors (PRRs), thereby initiating innate immune responses and subsequent adaptive immune responses. PAMPs can be developed as a vaccine adjuvant for modulating and optimizing antigen-specific immune responses, especially in combating viral infections and tumor therapy. Although several PAMP adjuvants have been successfully developed they are still lacking in general, and many of them are in the preclinical exploration stage. OBJECTIVE This review summarizes the research progress and development direction of PAMP adjuvants, focusing on their immune mechanisms and clinical applications. METHODS PubMed, Scopus, and Google Scholar were screened for this information. We highlight the immune mechanisms and clinical applications of PAMP adjuvants. RESULTS Because of the differences in receptor positions, specific immune cells targets, and signaling pathways, the detailed molecular mechanism and pharmacokinetic properties of one agonist cannot be fully generalized to another agonist, and each PAMP should be studied separately. In addition, combination therapy and effective integration of different adjuvants can increase the additional efficacy of innate and adaptive immune responses. CONCLUSION The mechanisms by which PAMPs exert adjuvant functions are diverse. With continuous discovery in the future, constant adjustments should be made to build new understandings. At present, the goal of therapeutic vaccination is to induce T cells that can specifically recognize and eliminate tumor cells and establish long-term immune memory. Following immune checkpoint modulation therapy, cancer treatment vaccines may be an option worthy of clinical testing.
Collapse
Affiliation(s)
- Yu Yan
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| | - Dan Yao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| | - Xiaoyu Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| |
Collapse
|
14
|
Zhou Y, Li M, Lv T, Huang M, Cheng B, Zhang Y, Zhu J. Gastrodin Inhibits Virus Infection by Promoting the Production of Type I Interferon. Front Pharmacol 2021; 11:608707. [PMID: 33776755 PMCID: PMC7990098 DOI: 10.3389/fphar.2020.608707] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/30/2020] [Indexed: 01/18/2023] Open
Abstract
Type I interferon (IFN-I) plays a critical role in the antiviral immune response. However, viruses have developed different strategies to suppress the production of IFN-I for its own escape and amplification. Therefore, promoting the production of IFN-I is an effective strategy against virus infection. Gastrodin (GTD), a phenolic glucoside extracted from Gastrodia elata Blume, has been reported to play a protective role in some central nervous system -related diseases and is beneficial for the recovery of diseases by inhibiting inflammation. However, the effect of GTD on virus infection is largely unknown. Here we found GTD treatment increased the survival rate of mice infected with vesicular stomatitis virus (VSV) or herpes simplex virus-1 (HSV-1). The production of IFN-I was increased in GTD-treated mice or macrophages compared to the control group, during virus infection. Furthermore, the activation of interferon regulatory factor 3 (IRF3) was promoted by GTD in macrophages upon VSV and HSV-1 infection. Our results demonstrated that GTD could inhibit the VSV and HSV-1 infection by promoting the production of IFN-I in macrophages and might provide an effective strategy against virus infection.
Collapse
Affiliation(s)
- Yunlian Zhou
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mengyao Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Tingyi Lv
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Meixia Huang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Beilei Cheng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuanyuan Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jie Zhu
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
15
|
Kuehling J, Eisenhofer K, Lechner M, Becker S, Willems H, Reiner G. The effects of boar on susceptibility to swine inflammation and necrosis syndrome in piglets. Porcine Health Manag 2021; 7:15. [PMID: 33509289 PMCID: PMC7842003 DOI: 10.1186/s40813-021-00194-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammation and necrosis can appear in pigs in several parts of the body simultaneously. The signs can affect newborns, suckling piglets and older pigs, and recent studies suggest that the syndrome is primarily endogenous. Inflammation and necrosis indicate impaired animal welfare, and thus should be controlled in pig production. This can be achieved by improving husbandry conditions. However, the variation in signs also appears to have a genetic component. The aim of the present study was therefore to test the effects of different boars from the Duroc and Pietrain breeds on the prevalence of swine inflammation and necrosis syndrome in their offspring. For this purpose, 646 suckling pigs from 39 sows (two herds) and 19 boars were made available. On the third day of life, the piglets were examined for clinical signs of inflammation and necrosis at tail base, tail tip, ears, face, teats, navel and claws. For the evaluation, we included the boar within the breed and the breed as fixed effects and the sow within the herd as random effects. More than 70% of the piglets were affected at the tail base, ears, coronary bands and heels. Bristle loss, swelling, redness, venous congestion and claw wall bleeding occurred most frequently. Exudation and necrosis affected fewer piglets. None of the piglets was completely free from signs of SINS. Offspring from Duroc boars had significantly lower SINS scores (4.87 ± 0.44) than offspring from Pietrain boars (10.13 ± 0.12). Within the Pietrain breed, significant effects of the boar were observed on inflammation and necrosis levels. Under the present study conditions, using Duroc boars instead of Pietrain boars resulted in a 59% reduction in the SINS scores of their offspring. The SINS score in the offspring of the most favourable Pietrain boar was almost 40% lower than that of offspring in the least favourable. These findings confirm considerable genetic effects on the outcome of SINS under a given husbandry. Further studies are necessary to characterise the genetic effects in detail and to make them useful to combat the syndrome.
Collapse
Affiliation(s)
- Josef Kuehling
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany
| | - Kathrin Eisenhofer
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany
| | | | - Sabrina Becker
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany
| | - Hermann Willems
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany
| | - Gerald Reiner
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany.
| |
Collapse
|
16
|
Gao X, Tang M, Tian S, Li J, Liu W. A ferroptosis-related gene signature predicts overall survival in patients with lung adenocarcinoma. Future Oncol 2021; 17:1533-1544. [PMID: 33432837 DOI: 10.2217/fon-2020-1113] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aims: To elucidate the association between ferroptosis-related genes and prognosis in patients with lung adenocarcinoma (LUAD). Materials & methods: A ferroptosis-related gene signature was made by lasso regression analysis through the LUAD datasets of the Cancer Genome Atlas. The prognostic value of the multigene signature was externally validated in the GSE72094 dataset from the Gene Expression Omnibus database. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analysis were used to explore underlying mechanisms. Results & conclusion: We established a novel ferroptosis-related gene signature for overall survival in LUAD that was predictive in both the training and validation cohorts. Immune-related pathways were significantly enriched, and immune status differed between the high- and low-risk groups. Targeting ferroptosis is a potential therapeutic option in LUAD. These results still need to be confirmed by more studies.
Collapse
Affiliation(s)
- Xinliang Gao
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Streat, Changchun, Jilin 130021, PR China
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Streat, Changchun, Jilin 130021, PR China
| | - Suyan Tian
- Division of Clinical Research, The First Hospital of Jilin University, 71 Xinmin Streat, Changchun, Jilin 130021, PR China
| | - Jialin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Streat, Changchun, Jilin 130021, PR China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Streat, Changchun, Jilin 130021, PR China
| |
Collapse
|
17
|
Ding J, Liu X, Tang B, Bai X, Wang Y, Li S, Li J, Liu M, Wang X. Murine hepatoma treatment with mature dendritic cells stimulated by Trichinella spiralis excretory/secretory products. Parasite 2020; 27:47. [PMID: 32692308 PMCID: PMC7373160 DOI: 10.1051/parasite/2020045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 07/03/2020] [Indexed: 11/14/2022] Open
Abstract
Excretory/Secretory Products (ESPs) of the nematode Trichinella spiralis contain antitumor-active substances that inhibit tumor growth. Mature dendritic cells (DCs) play a critical role in the antitumor immunity of the organism. As pathogen-derived products, it ought to be discussed whether T. spiralis ESPs will reduce the antitumor effect of mature DCs from the host before it is applied to patients' tumors. Therefore, the aim of this work was to evaluate the immunological effect of DCs stimulated by T. spiralis ESPs in H22 tumor-bearing mice. H22 tumor model mice in this study were randomly divided into four groups according to the treatment: PBS control group, ESP group, DCs group, and DCs stimulated with T. spiralis ESP (ESP+DCs group). The antitumor effect was evaluated by tumor inhibition rate and cytokine detection using ELISA. The results showed significant inhibition in tumor growth in the ESP+DCs, DCs and ESP groups when compared with the PBS control group (p < 0.01, p < 0.01, and p < 0.05, respectively). However, no significant difference was observed on tumor inhibition rates between the ESP+DCs and DCs groups. The decrease in IL-4, IL-6, and IL-10, and the increase in IFN-γ between the DCs and ESP+DCs groups were also not significant. Therefore, DCs stimulated by ESP did not reduce the antitumor effect of mature DCs, which demonstrated that the T. spiralis ESP would not affect the antitumor effect of mature DCs by modulating the immune response of the host, and that ESPs are safe in antitumor immunology when applied in a tumor model mice.
Collapse
Affiliation(s)
- Jing Ding
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Xiaolei Liu
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Bin Tang
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Xue Bai
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Yang Wang
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Shicun Li
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Jian Li
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Mingyuan Liu
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| | - Xuelin Wang
- Key Laboratory for Zoonoses Research, Ministry of Education, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in Asian-Pacific Region Changchun 130062 P.R. China
| |
Collapse
|
18
|
Zhou Y, Li M, Lv T, Huang M, Cheng B, Zhang Y, Zhu J. Gastrodin Inhibits Virus Infection by Promoting the Production of Type I Interferon. Front Pharmacol 2020. [PMID: 33776755 DOI: 10.3389/fphar.2020.00931/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Type I interferon (IFN-I) plays a critical role in the antiviral immune response. However, viruses have developed different strategies to suppress the production of IFN-I for its own escape and amplification. Therefore, promoting the production of IFN-I is an effective strategy against virus infection. Gastrodin (GTD), a phenolic glucoside extracted from Gastrodia elata Blume, has been reported to play a protective role in some central nervous system -related diseases and is beneficial for the recovery of diseases by inhibiting inflammation. However, the effect of GTD on virus infection is largely unknown. Here we found GTD treatment increased the survival rate of mice infected with vesicular stomatitis virus (VSV) or herpes simplex virus-1 (HSV-1). The production of IFN-I was increased in GTD-treated mice or macrophages compared to the control group, during virus infection. Furthermore, the activation of interferon regulatory factor 3 (IRF3) was promoted by GTD in macrophages upon VSV and HSV-1 infection. Our results demonstrated that GTD could inhibit the VSV and HSV-1 infection by promoting the production of IFN-I in macrophages and might provide an effective strategy against virus infection.
Collapse
Affiliation(s)
- Yunlian Zhou
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mengyao Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Tingyi Lv
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Meixia Huang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Beilei Cheng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuanyuan Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jie Zhu
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
19
|
Vicente D, Patino M, Marcus R, Lillmoe H, Limani P, Newhook T, Lee A, Tzeng CW, Segraves-Chun Y, Tweardy D, Gottumukkala V, Vauthey JN, Aloia T, Cata JP. Impact of epidural analgesia on the systemic biomarker response after hepatic resection. Oncotarget 2019; 10:584-594. [PMID: 30728909 PMCID: PMC6355178 DOI: 10.18632/oncotarget.26549] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/22/2018] [Indexed: 12/30/2022] Open
Abstract
Background Perioperative inflammation is associated with poor oncologic outcomes. Regional analgesia has been shown mitigate some of these inflammatory changes and be associated with better oncologic outcomes in patients with hepatic malignancies. The mechanism for this effect, however, remains unclear. The authors sought to compare systemic biomarker concentrations in a comprehensive and oncologically relevant panel in the perioperative setting between patients undergoing thoracic epidural analgesia (TEA) and intra-venous patient- controlled analgesia (IV-PCA) for resection of hepatic metastatic disease. Results Clinicopathologic variables and baseline biomarkers were similar between TEA (n = 46) and IV-PCA (n = 16) groups. Of the biomarkers which were significantly changed from baseline, there was a lower fold change from baseline in the TEA patients compared to IV-PCA including IL-6 (13.5vs19.1), MCP-1 (1.9vs3.0), IL-8 (2.4vs3.0), and Pentraxin-3 (10.8vs15.6). Overall decreased systemic concentrations of TGFb signaling were noted in TEA patients on POD1 TGFb3 (243.2 vs. 86.0, p = 0.005), POD3 TGFb1 (6558.0 vs. 2063.3, p = 0.004), POD3 TGFb2 (468.3 vs. 368.9, p = 0.036), POD3 TGFb3 (132.2 vs. 77.8, p = 0.028), and POD5 TGFb3 (306.5 vs. 92.2, p = 0.032). POD1 IL-12p70 concentrations were significantly higher in TEA patients (8.3 vs. 1.6, p = 0.024). Conclusion Epidural analgesia damped the postoperative inflammatory response and systemic immunosuppressive signaling, as well as promoted Th1 systemic signaling early in the post-operative period after hepatic resection for metastatic disease. These differences elaborate on known mechanisms for improved oncologic outcomes with regional anesthesia, and may be considered for biomarker monitoring of effective regional anesthesia in oncologic surgery. Materials and Methods Patient data, including clinicopathologic variables were collected for this study from the database of a randomized controlled trial comparing perioperative outcomes in patients undergoing hepatic resection with TEA vs. IV-PCA. Patients undergoing resection for metastatic disease were selected for this study. Plasma concentrations (pg/mL) of well-studied biomarkers (IL-1b/2/4/5/6/7/8/10/12p70/13/17, MCP-1 IFNγ, TNFα, MIP-1b, GM-CSF, G-CSF, VEGF, Resistin, TGFb1, TGFb2, and TGFb3), as well as novel perioperative markers (CXCL12, CXCL10, Omentin-1, sLeptin R, Vaspin, Pentraxin-3, Galactin-3, FGF-23, PON-1, FGF-21) were measured preoperatively, and on postoperative day (POD)1, POD3, and POD5 using multiplex bead assays. Clinicopathologic variables and perioperative variations in these biomarkers were compared between TEA vs IV-PCA groups.
Collapse
Affiliation(s)
- Diego Vicente
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Miguel Patino
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.,Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca Marcus
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Lillmoe
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Preparim Limani
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy Newhook
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andy Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yun Segraves-Chun
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Tweardy
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vijaya Gottumukkala
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Aloia
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan P Cata
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.,Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
Laskarin G, Gulic T, Glavan Gacanin L, Dominovic M, Haller H, Rukavina D. Assessing whether progesterone-matured dendritic cells are responsible for retention of fertilization products in missed abortion. Med Hypotheses 2018; 118:169-173. [DOI: 10.1016/j.mehy.2018.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/01/2018] [Accepted: 04/11/2018] [Indexed: 12/25/2022]
|
21
|
El-Murr T, Patel A, Sedlak C, D'Souza-Lobo B. Evaluating dendritic cells as an in vitro screening tool for immunotherapeutic formulations. J Immunol Methods 2018; 459:55-62. [PMID: 29800576 DOI: 10.1016/j.jim.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/09/2018] [Accepted: 05/09/2018] [Indexed: 01/24/2023]
Abstract
Immunotherapy approaches targeting dendritic cells (DCs) are being studied as treatment options in cancer. This project focused on utilizing DCs as a valuable in vitro screening tool for efficacious microparticle formulations containing tumor associated antigens (TAAs) and adjuvants as immunotherapy alternatives. The innate immune system, including DCs, distinctly responds to the particulate matter and adjuvants in these formulations which stimulates the adaptive immune system to eliminate resident cancer cells. We formulated microparticles (MPs) co-loaded with TAAs along with the adjuvants, AddaVax™ and Imiquimod, and measured their effect on DCs in eliciting a cell-mediated immune response towards tumors. The MP zeta potential was measured as -24.0 mV and -26.5 mV for blank and TAA/adjuvant co-loaded microparticles, and the average particle size was 671.2 nm and 854.4 nm respectively. We determined that nitric oxide (NO) secretion was significantly higher in the adjuvant MP treated DCs group and was dose dependent with 1 mg/mL demonstrating the highest secretion levels. TNF-α release was highest in AddaVax™/TAA and Imiquimod/TAA MPs treated DCs, while IL-6 secretion was highest from Imiquimod/TAA MPs as well as from combined AddaVax™/TAA and Imiquimod/TAA MPs. Overall, the cell surface marker expressions of CD80, CD86, CD40, CD54, MHC-I and MHC-II levels were highest with combined AddaVax™/TAA and Imiquimod/TAA MPs. The results of our experiments suggest that a combination of adjuvants targeting different DC receptors loaded with TAA MPs creates an efficient delivery system to T-cells that could improve adaptive immune responses. Our studies also confirm that DCs are potent innate immune cells that can be used successfully as an in vitro tool to screen novel delivery formulations focused on immunotherapy.
Collapse
Affiliation(s)
- Theresa El-Murr
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States
| | - Ankita Patel
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States
| | - Carrie Sedlak
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States
| | - Bernadette D'Souza-Lobo
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States.
| |
Collapse
|
22
|
Significance of Frequencies, Compositions, and/or Antileukemic Activity of (DC-stimulated) Invariant NKT, NK and CIK Cells on the Outcome of Patients With AML, ALL and CLL. J Immunother 2018; 40:224-248. [PMID: 28557814 DOI: 10.1097/cji.0000000000000171] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Invariant natural killer T (iNKT)/natural killer (NK)/cytokine-induced killer (CIK) cells are important for immune surveillance. (I) Novel combinations of antibody 6B11 (targeting the Vα24-Jα18-invariant T-cell receptor) with CD4/CD8/CD1d/Vα24 for iNKT subset detection and "T/NK cell-like"-iNKT subsets were defined. Compared with healthy peripheral blood mononuclear cells (MNC) (significantly) lower proportions of iNKT cells (6B11/6B11CD3/6B11CD161), NK cells (CD3CD56/CD3CD161), and CIK cells (CD3CD56/CD3CD161) were found in peripheral blood MNC from acute myeloid (AML)/acute myeloid, lymphoid (ALL)/chronic lymphoid leukemia (CLL) patients in acute disease stages. Subtyping of iNKT cells revealed (significantly) higher proportions of CD3 T cells and CD161 NK cells in AML/ALL/CLL expressing 6B11 compared with healthy MNC. Prognostic evaluations showed higher proportions of iNKT/NK/CIK cells in favorable AML subgroups (younger age, primary, no extramedullary disease, achievement/maintenance of complete remission) or adult ALL and CLL patients. (II) iNKT/NK/CIK cell frequencies increased after (vs. before) mixed lymphocyte cultures of T-cell-enriched immune reactive cells stimulated with MNC/whole blood with or without pretreatment with "cocktails" (dendritic cells generating methods/kits inducing blasts' conversion to leukemia-derived dendritic cells from AML patients). Individual "cocktails" leading to "highest" iNKT cell frequencies could be defined. Antileukemic blast lytic activity correlated significantly with frequencies of iNKT/NK/CIK cells. In summary healthy MNC show significantly more iNKT/NK/CIK cells compared with AML/ALL/CLL MNC, a shift in the iNKT cell composition is seen in healthy versus leukemic samples and iNKT/NK/CIK cell-proportions in AML/ALL/CLL MNC samples correlate with prognosis. "Cocktail"-treated AML blasts lead to higher iNKT/NK/CIK cell frequencies and samples with antileukemic activity show significantly higher frequencies of iNKT/NK/CIK cells. Proportions of iNKT/NK/CIK cells should regularly be evaluated in AML/ALL/CLL diagnosis panels for quantitative/prognostic estimation of individual patients' antileukemic potential and their role in dendritic cells/leukemia-derived dendritic cells triggered immune surveillance.
Collapse
|
23
|
Ngu LN, Nji NN, Ambada G, Ngoh AA, Njambe Priso GD, Tchadji JC, Lissom A, Magagoum SH, Sake CN, Tchouangueu TF, Chukwuma GO, Okoli AS, Sagnia B, Chukwuanukwu R, Tebit DM, Esimone CO, Waffo AB, Park CG, Überla K, Nchinda GW. Dendritic cell targeted HIV-1 gag protein vaccine provides help to a recombinant Newcastle disease virus vectored vaccine including mobilization of protective CD8 + T cells. IMMUNITY INFLAMMATION AND DISEASE 2017; 6:163-175. [PMID: 29205929 PMCID: PMC5818444 DOI: 10.1002/iid3.209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022]
Abstract
Introduction Recombinant Newcastle Disease virus (rNDV) vectored vaccines are safe mucosal applicable vaccines with intrinsic immune‐modulatory properties for the induction of efficient immunity. Like all viral vectored vaccines repeated inoculation via mucosal routes invariably results to immunity against viral vaccine vectors. To obviate immunity against viral vaccine vectors and improve the ability of rNDV vectored vaccines in inducing T cell immunity in murine air way we have directed dendritic cell targeted HIV‐1 gag protein (DEC‐Gag) vaccine; for the induction of helper CD4+ T cells to a Recombinant Newcastle disease virus expressing codon optimized HIV‐1 Gag P55 (rNDV‐L‐Gag) vaccine. Methods We do so through successive administration of anti‐DEC205‐gagP24 protein plus polyICLC (DEC‐Gag) vaccine and rNDV‐L‐Gag. First strong gag specific helper CD4+ T cells are induced in mice by selected targeting of anti‐DEC205‐gagP24 protein vaccine to dendritic cells (DC) in situ together with polyICLC as adjuvant. This targeting helped T cell immunity develop to a subsequent rNDV‐L‐Gag vaccine and improved both systemic and mucosal gag specific immunity. Results This sequential DEC‐Gag vaccine prime followed by an rNDV‐L‐gag boost results to improved viral vectored immunization in murine airway, including mobilization of protective CD8+ T cells to a pathogenic virus infection site. Conclusion Thus, complementary prime boost vaccination, in which prime and boost favor distinct types of T cell immunity, improves viral vectored immunization, including mobilization of protective CD8+T cells to a pathogenic virus infection site such as the murine airway.
Collapse
Affiliation(s)
- Loveline N Ngu
- Department of Biochemistry, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon.,Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon
| | - Nadesh N Nji
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon
| | - Georgia Ambada
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Apeh A Ngoh
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of biomedical sciences, University of Dschang, Dschang, Cameroon
| | - Ghislain D Njambe Priso
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Jules C Tchadji
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Abel Lissom
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Suzanne H Magagoum
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Carol N Sake
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Microbiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Thibau F Tchouangueu
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of biochemistry, University of Dschang, Dschang, Cameroon
| | - George O Chukwuma
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Medical Laboratory Science College of Medicine, Nnewi Campus, Nnamdi Azikiwe University, Awka, Anambra
| | | | - Bertrand Sagnia
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon
| | - Rebecca Chukwuanukwu
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Medical Laboratory Science College of Medicine, Nnewi Campus, Nnamdi Azikiwe University, Awka, Anambra
| | - Denis M Tebit
- Myles Thaler Center for AIDS and Human Retrovirus Research, Department of Microbiology, Immunology and Cancer Biology, Jordan Hall 7088, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22903, USA
| | - Charles O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| | - Alain B Waffo
- Department of Biological Sciences # 223, Alabama State University, 1627, Hall Street, Montgomery, Alabama 36104, USA
| | - Chae G Park
- Laboratory of Immunology, Brain Korea 21 PLUS Project for Medical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York 10065, USA
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Erlangen, Germany
| | - Godwin W Nchinda
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
24
|
Abstract
Safe and efficacious vaccines are arguably the most successful medical interventions of all time. Yet the ongoing discovery of new pathogens, along with emergence of antibiotic-resistant pathogens and a burgeoning population at risk of such infections, imposes unprecedented public health challenges. To meet these challenges, innovative strategies to discover and develop new or improved anti-infective vaccines are necessary. These approaches must intersect the most meaningful insights into protective immunity and advanced technologies with capabilities to deliver immunogens for optimal immune protection. This goal is considered through several recent advances in host-pathogen relationships, conceptual strides in vaccinology, and emerging technologies. Given a clear and growing risk of pandemic disease should the threat of infection go unmet, developing vaccines that optimize protective immunity against high-priority and antibiotic-resistant pathogens represents an urgent and unifying imperative.
Collapse
Affiliation(s)
- Michael R Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90024.,Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509; .,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509.,Los Angeles Biomedical Research Institute, Torrance, California 90502
| | | |
Collapse
|
25
|
Bazzi S, Modjtahedi H, Mudan S, Achkar M, Akle C, Bahr GM. Immunomodulatory effects of heat-killed Mycobacterium obuense on human blood dendritic cells. Innate Immun 2017; 23:592-605. [PMID: 28853313 DOI: 10.1177/1753425917727838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Heat-killed (HK) Mycobacterium obuense is a novel immunomodulator, currently undergoing clinical evaluation as an immunotherapeutic agent in the treatment of cancer. Here, we examined the effect of in vitro exposure to HK M. obuense on the expression of different categories of surface receptors on human blood myeloid (m) and plasmacytoid (p) DCs. Moreover, we have characterized the cytokine and chemokine secretion patterns of purified total blood DCs stimulated with HK M. obuense. HK M. obuense significantly up-regulated the expression of CD11c, CD80, CD83, CD86, CD274 and MHC class II in whole-blood mDCs and CD80, CD123 and MHC class II in whole-blood pDCs. Down-regulation of CD195 expression in both DC subpopulations was also noted. Further analysis showed that HK M. obuense up-regulated the expression of CD80, CD83 and MHC class II on purified blood DC subpopulations. TLR2 and TLR1 were also identified to be engaged in mediating the HK M. obuense-induced up-regulation of surface receptor expression on whole blood mDCs. In addition, our data demonstrated that HK M. obuense augmented the secretion of CCL4, CCL5, CCL22, CXCL8, IL-6, IL-12p40 and TNF-α by purified total blood DCs. Taken together, our data suggest that HK M. obuense exerts potent differential immunomodulatory effects on human DC subpopulations.
Collapse
Affiliation(s)
- Samer Bazzi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK.,2 Biology Department, Faculty of Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| | - Helmout Modjtahedi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK
| | - Satvinder Mudan
- 3 St George's University of London, Imperial College, London and The Royal Marsden Hospital, London, UK
| | - Marcel Achkar
- 4 Clinical Laboratory Department, Nini Hospital, Tripoli, Lebanon
| | | | - Georges M Bahr
- 6 Faculty of Medicine and Medical Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| |
Collapse
|