1
|
Singh S, Verma P, Gaur M, Bhati L, Madan R, Sharma PP, Rawat A, Rathi B, Singh M. In-silico development of a novel TLR2-mediating multi-epitope vaccine against Mycobacterium tuberculosis. In Silico Pharmacol 2025; 13:34. [PMID: 40018380 PMCID: PMC11861476 DOI: 10.1007/s40203-025-00322-8] [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: 11/28/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), still remains one of the leading causes of mortality worldwide. The elusive nature of this pathogen and its ability to develop drug resistance makes it a serious threat to global health. BCG, the only preventive vaccine for TB, has a limited efficacy and provides partial protection against the disease. A new effective recombinant vaccine capable of producing a stronger and more comprehensive immune response is required to address this global threat. In the present study, we adopted an in-silico approach to develop a multi-epitope vaccine by screening 198 "regulatory proteins" of Mtb H37Rv strain. Epitopes generated from these proteins were screened on the basis of antigenicity, cytokine profile, allergenicity, toxicity, conservancy and population coverage. Selected epitopes were docked with predominant MHC alleles that were used to develop a vaccine construct using suitable linkers and adjuvant. The construct was subjected to homology modelling, tertiary structure validation and refinement and was eventually docked with Toll-like receptor 2 receptor. Molecular dynamic simulation studies revealed stable interactions between the vaccine construct and TLR-2 receptor. The construct also displayed a high probability to elicit a protective immune response involving both humoral and cell-mediated components. In conclusion, the findings suggest that the constructed vaccine has the potential to induce a robust immune response against Mtb. However, further in-vitro and in-vivo studies are required to assess the safety, efficacy, and long-term protective effects of the vaccine construct. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-025-00322-8.
Collapse
Affiliation(s)
- Swati Singh
- Department of Zoology, University of Delhi, Delhi, India
| | - Priyanshu Verma
- Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany
- Hansraj College, University of Delhi, New Delhi, India
| | - Madhav Gaur
- School of Life Sciences, Technical University of Munich, Munich, Germany
- Hansraj College, University of Delhi, New Delhi, India
| | - Lavi Bhati
- Department of Reproductive Biology, All India Institute for Medical Sciences, New Delhi, India
- Hansraj College, University of Delhi, New Delhi, India
| | - Riya Madan
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
- Hansraj College, University of Delhi, New Delhi, India
| | - Prem P. Sharma
- Hansraj College, University of Delhi, New Delhi, India
- HeteroChem InnoTech, New Delhi, Delhi India
| | - Ayushi Rawat
- Hansraj College, University of Delhi, New Delhi, India
| | - Brijesh Rathi
- H.G. Khorana Centre for Chemical Biology, Department of Chemistry, Hansraj College, University of Delhi, New Delhi, India
| | - Medha Singh
- Centre for Tuberculosis Research, School of Medicine, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
2
|
Cardoso KF, de Souza LRA, da Silva Santos BSÁ, de Carvalho KRA, da Silva Messias SG, de Faria Gonçalves AP, Kano FS, Alves PA, da Silva Campos MA, Xavier MP, Garcia CC, Russo RC, Gazzinelli RT, Costa ÉA, da Silva Martins NR, Miyaji EN, de Magalhães Vieira Machado A, Silva Araújo MS. Intranasal influenza-vectored vaccine expressing pneumococcal surface protein A protects against Influenza and Streptococcus pneumoniae infections. NPJ Vaccines 2024; 9:246. [PMID: 39702744 DOI: 10.1038/s41541-024-01033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
Streptococcus pneumoniae and influenza A virus (IAV) are significant agents of pneumonia cases and severe respiratory infections globally. Secondary bacterial infections, particularly by Streptococcus pneumoniae, are common in IAV-infected individuals, leading to critical outcomes. Despite reducing mortality, pneumococcal vaccines have high production costs and are serotype specific. The emergence of new circulating serotypes has led to the search for new prevention strategies that provide a broad spectrum of protection. In this context, vaccination using antigens present in all serotypes, such as Pneumococcal Surface Protein A (PspA), can offer broad coverage regardless of serotype. Employing the reverse genetics technique, our research group developed a recombinant influenza A H1N1 virus that expresses PspA (Flu-PspA), through the replacement of neuraminidase by PspA. This virus was evaluated as a bivalent vaccine against infections caused by influenza A and S. pneumoniae in mice. Initially, we evaluated the Flu-PspA virus's ability to infect cells and express PspA in vitro, its capacity to multiply in embryonated chicken eggs, and its safety when inoculated in mice. Subsequently, the protective effect against influenza A and Streptococcus pneumoniae lethal challenge infections in mice was assessed using different immunization protocols. Analysis of the production of antibodies against PspA4 protein and influenza, and the binding capacity of anti-PspA4 antibodies/complement deposition to different strains of S. pneumoniae were also evaluated. Our results demonstrate that the Flu-PspA virus vaccine efficiently induces PspA protein expression in vitro, and that it was able to multiply in embryonated chicken eggs even without exogenous neuraminidase. The Flu-PspA-based bivalent vaccine was demonstrated to be safe, stimulated high titers of anti-PspA and anti-influenza antibodies, and protected mice against homosubtypic and heterosubtypic influenza A and S. pneumoniae challenge. Moreover, an efficient binding of antibodies and complement deposition on the surface of pneumococcal strains ascribes the broad-spectrum vaccine response in vivo. In summary, this innovative approach holds promise for developing a dual-protective vaccine against two major respiratory pathogens.
Collapse
Affiliation(s)
- Kimberly Freitas Cardoso
- Laboratório de Imunologia de Doenças Virais, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Lara Regina Alves de Souza
- Laboratório de Imunologia de Doenças Virais, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | | | | | - Sarah Giarola da Silva Messias
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Ana Paula de Faria Gonçalves
- Laboratório de Imunologia de Doenças Virais, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Flora Satiko Kano
- Grupo de Pesquisa em Biologia Molecular e Imunologia da Malária, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Pedro Augusto Alves
- Laboratório de Imunologia de Doenças Virais, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Marco Antônio da Silva Campos
- Laboratório de Imunologia de Doenças Virais, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Marcelo Pascoal Xavier
- Laboratório de Imunologia de Doenças Virais, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Cristiana Couto Garcia
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Remo Castro Russo
- Laboratório de Imunologia e Mecânica Pulmonar, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Ricardo Tostes Gazzinelli
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Érica Azevedo Costa
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | | | | | | | - Márcio Sobreira Silva Araújo
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou-Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil.
| |
Collapse
|
3
|
Fernandes-Braga W, Curotto de Lafaille MA. B cell memory of Immunoglobulin E (IgE) antibody responses in allergy. Curr Opin Immunol 2024; 91:102488. [PMID: 39340881 DOI: 10.1016/j.coi.2024.102488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Immunoglobulin E (IgE)-mediated allergic diseases are driven by high-affinity allergen-specific IgE antibodies. IgE antibodies bind to Fc epsilon receptors on mast cells, prompting their degranulation and initiating inflammatory reactions upon allergen crosslinking. While most IgE-producing plasma cells have short lifespans, and IgE memory B cells are exceedingly rare, studies have indicated that non-IgE-expressing type 2-polarized IgG memory B cells serve as a reservoir of IgE memory in allergies. This review explores the B cell populations underlying IgE-mediated allergies, including the cellular and molecular processes that drive IgE class switching from non-IgE memory B cells. It highlights emerging evidence from human studies identifying type 2 IgG memory B cells as the source of pathogenic IgE in allergic responses.
Collapse
Affiliation(s)
- Weslley Fernandes-Braga
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, and Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria A Curotto de Lafaille
- Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, and Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Ding D, Gao R, Lei Y, Liu J, Zhou C, Wen Y, Zhou S, Guo J, Li T. Synergistic immune augmentation enabled by covalently conjugating TLR4 and NOD2 agonists. Eur J Med Chem 2024; 278:116792. [PMID: 39217861 DOI: 10.1016/j.ejmech.2024.116792] [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: 07/09/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Enhancing the efficacy of subunit vaccines relies significantly on the utilization of potent adjuvants, particularly those capable of triggering multiple immune pathways. To achieve synergistic immune augmentation by Toll-like receptor 4 agonist (TLR4a) and nucleotide-binding oligomerization-domain-containing protein 2 agonist (NOD2a), in this work, we conjugated RC529 (TLR4a) and MDP (NOD2a) to give RC529-MDP, and evaluated its adjuvanticity for OVA antigen. Compared to the unconjugated RC529+MDP, RC529-MDP remarkably enhanced innate immune responses with 6.8-fold increase in IL-6 cytokine, and promoted the maturation of antigen-presenting cells (APCs), possibly because of the conjugation of multiple agonists ensuring their delivery to the same cell and activation of various signaling pathways within that cell. Furthermore, RC529-MDP improved OVA-specific antibody response, T cells response and the memory T cells ratio relative to the unconjugated mixture. Therefore, covalently conjugating TLR4 agonist and NOD2 agonist was an effective strategy to enhance immune responses, providing the potential to design and develop more effective vaccines.
Collapse
Affiliation(s)
- Dong Ding
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Runing Gao
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yujuan Lei
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Jianing Liu
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chengkai Zhou
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yu Wen
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Shihao Zhou
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Jun Guo
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Tiehai Li
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
5
|
Gao L, Li G, Qiu C, Ye Y, Li X, Liao P, Ming W, Liu Z, Luo X, Liao G. Design, Synthesis, and Bioactivity Evaluation of a TF-Based Cancer Vaccine Candidate Using Lipid A Mimetics As a Built-In Adjuvant. J Med Chem 2024; 67:9976-9990. [PMID: 38886162 DOI: 10.1021/acs.jmedchem.4c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
This study describes the design and synthesis of five TF-based cancer vaccine candidates using a lipid A mimetic as the carrier and a built-in adjuvant. All synthesized conjugates elicited robust and consistent TF-specific immune responses in mice without external adjuvants. Immunological studies subsequently conducted in wild-type and TLR4 knockout C57BL/6 mice demonstrated that the activation of TLR4 was the main reason that the synthesized lipid A mimetics increased the TF-specific immune responses. All antisera induced by these conjugates can specifically recognize, bind to, and induce the lysis of TF-positive cancer cells. Moreover, representative conjugates 2 and 3 could effectively reduce the growth of tumors and prolong the survival time of mice in vivo, and the efficacies were better than glycoprotein TF-CRM197 with alum adjuvant. Lipid A mimetics could therefore be a promising platform for the development of new carbohydrate-based vaccine carriers with self-adjuvanting properties for the treatment of cancer.
Collapse
Affiliation(s)
- Lingqiang Gao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guiqi Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Cuiping Qiu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yifan Ye
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaohui Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Pan Liao
- Guangzhou Yuemei Pharmaceutical Technology Co., Ltd, Guangzhou 510535, China
| | - Wenbo Ming
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiang Luo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guochao Liao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
6
|
Zheng X, Yang R, Zhao Y, Zhang Y, Yuan G, Li W, Xiao Z, Dong X, Ma M, Guo Y, Wang W, Zhao X, Yang H, Qiu S, Peng Z, Liu A, Yu S, Zhang Y. Alum/CpG adjuvant promotes immunogenicity of inactivated SARS-CoV-2 Omicron vaccine through enhanced humoral and cellular immunity. Virology 2024; 594:110050. [PMID: 38479071 DOI: 10.1016/j.virol.2024.110050] [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: 08/21/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 04/09/2024]
Abstract
The SARS-CoV-2 Omicron variant, which was classified as a variant of concern (VOC) by the World Health Organization on 26 November 2021, has attracted worldwide attention for its high transmissibility and immune evasion ability. The existing COVID-19 vaccine has been shown to be less effective in preventing Omicron variant infection and symptomatic infection, which brings new challenges to vaccine development and application. Here, we evaluated the immunogenicity and safety of an Omicron variant COVID-19 inactivated vaccine containing aluminum and CpG adjuvants in a variety of animal models. The results showed that the vaccine candidate could induce high levels of neutralizing antibodies against the Omicron variant virus and binding antibodies, and significantly promoted cellular immune response. Meanwhile, the vaccine candidate was safe. Therefore, it provided more foundation for the development of aluminum and CpG as a combination adjuvant in human vaccines.
Collapse
Affiliation(s)
- Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Rong Yang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yuxiu Zhao
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yadan Zhang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Guangying Yuan
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Weidong Li
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Zhuangzhuang Xiao
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Xiaofei Dong
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Meng Ma
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Yancen Guo
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Wei Wang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Xue Zhao
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Hongqiang Yang
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Shaoting Qiu
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Zheng Peng
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Ankang Liu
- Beijing Institute of Biological Products Company Limited, Beijing, China
| | - Shouzhi Yu
- Beijing Institute of Biological Products Company Limited, Beijing, China.
| | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing, China; China National Biotec Group Company Limited, Beijing, China.
| |
Collapse
|
7
|
Oser L, Midha A, Schlosser-Brandenburg J, Rausch S, Mugo RM, Kundik A, Elizalde-Velázquez LE, Adjah J, Musimbi ZD, Klopfleisch R, Helm CS, von Samson-Himmelstjerna G, Hartmann S, Ebner F. Ascaris suum infection in juvenile pigs elicits a local Th2 response in a setting of ongoing Th1 expansion. Front Immunol 2024; 15:1396446. [PMID: 38799456 PMCID: PMC11116563 DOI: 10.3389/fimmu.2024.1396446] [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: 03/05/2024] [Accepted: 04/08/2024] [Indexed: 05/29/2024] Open
Abstract
Ascaris spp. undergo extensive migration within the body before establishing patent infections in the small intestinal tract of humans and pigs. However, whether larval migration is critical for inducing efficient type 2 responses remains poorly understood. Therefore, we investigated systemic versus local adaptive immune responses along the hepato-tracheal migration of Ascaris suum during primary, single infections in conventionally raised pigs. Neither the initial invasion of gut tissue nor migration through the liver resulted in discernable Th2 cell responses. In contrast, lung-stage larvae elicited a Th2-biased pulmonary response, which declined after the larvae had left the lungs. In the small intestine, we observed an accumulation of Th2 cells upon the arrival of fourth-stage larvae (L4) to the small intestinal lumen. In parallel, we noticed robust and increasing Th1 responses in circulation, migration-affected organs, and draining lymph nodes. Phenotypic analysis of CD4+ T cells specifically recognizing A. suum antigens in the circulation and lung tissue of infected pigs confirmed that the majority of Ascaris-specific T cells produced IL-4 (Th2) and, to a much lesser extent, IL-4/IFN-g (Th2/1 hybrids) or IFN-g alone (Th1). These data demonstrate that lung-stage but not the early liver-stage larvae lead to a locally restricted Th2 response. Significant Th2 cell accumulation in the small intestine occurs only when L4 complete the body migration. In addition, Th2 immunity seems to be hampered by the concurrent, nonspecific Th1 bias in growing pigs. Together, the late onset of Th2 immunity at the site of infection and the Th1-biased systemic immunity likely enable the establishment of intestinal infections by sufficiently large L4 stages and pre-adult worms, some of which resist expulsion mechanisms.
Collapse
Affiliation(s)
- Larissa Oser
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Ankur Midha
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Josephine Schlosser-Brandenburg
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Rausch
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Robert M. Mugo
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Arkadi Kundik
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Luis E. Elizalde-Velázquez
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Joshua Adjah
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Zaneta D. Musimbi
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Christina S. Helm
- Department of Veterinary Medicine, Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Georg von Samson-Himmelstjerna
- Department of Veterinary Medicine, Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Susanne Hartmann
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Friederike Ebner
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
- Infection Pathogenesis, School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
8
|
Singh V, Nandi S, Ghosh A, Adhikary S, Mukherjee S, Roy S, Das C. Epigenetic reprogramming of T cells: unlocking new avenues for cancer immunotherapy. Cancer Metastasis Rev 2024; 43:175-195. [PMID: 38233727 DOI: 10.1007/s10555-024-10167-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024]
Abstract
T cells, a key component of cancer immunotherapy, undergo a variety of histone modifications and DNA methylation changes since their bone marrow progenitor stages before developing into CD8+ and CD4+ T cells. These T cell types can be categorized into distinct subtypes based on their functionality and properties, such as cytotoxic T cells (Tc), helper T cells (Th), and regulatory T cells (Treg) as subtypes for CD8+ and CD4+ T cells. Among these, the CD4+ CD25+ Tregs potentially contribute to cancer development and progression by lowering T effector (Teff) cell activity under the influence of the tumor microenvironment (TME). This contributes to the development of therapeutic resistance in patients with cancer. Subsequently, these individuals become resistant to monoclonal antibody therapy as well as clinically established immunotherapies. In this review, we delineate the different epigenetic mechanisms in cancer immune response and its involvement in therapeutic resistance. Furthermore, the possibility of epi-immunotherapeutic methods based on histone deacetylase inhibitors and histone methyltransferase inhibitors are under investigation. In this review we highlight EZH2 as the principal driver of cancer cell immunoediting and an immune escape regulator. We have addressed in detail how understanding T cell epigenetic regulation might bring unique inventive strategies to overcome drug resistance and increase the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Aritra Ghosh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Indian Institute of Science Education and Research, Kolkata, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Shravanti Mukherjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Siddhartha Roy
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India.
- Homi Bhabha National Institute, Mumbai, 400094, India.
| |
Collapse
|
9
|
Zimmermann J, van Haren SD, Diray-Arce J, Adriawan IR, Wørzner K, Krog RT, Guleed S, Hu T, Mortensen R, Dietrich J, Solbak SMØ, Levy O, Christensen D, Pedersen GK. Co-adjuvanting DDA/TDB liposomes with a TLR7 agonist allows for IgG2a/c class-switching in the absence of Th1 cells. NPJ Vaccines 2023; 8:189. [PMID: 38135685 PMCID: PMC10746746 DOI: 10.1038/s41541-023-00781-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Class-switching to IgG2a/c in mice is a hallmark response to intracellular pathogens. T cells can promote class-switching and the predominant pathway for induction of IgG2a/c antibody responses has been suggested to be via stimulation from Th1 cells. We previously formulated CAF®01 (cationic liposomes containing dimethyldioctadecylammonium bromide (DDA) and Trehalose-6,6-dibehenate (TDB)) with the lipidated TLR7/8 agonist 3M-052 (DDA/TDB/3M-052), which promoted robust Th1 immunity in newborn mice. When testing this adjuvant in adult mice using the recombinant Chlamydia trachomatis (C.t.) vaccine antigen CTH522, it similarly enhanced IgG2a/c responses compared to DDA/TDB, but surprisingly reduced the magnitude of the IFN-γ+Th1 response in a TLR7 agonist dose-dependent manner. Single-cell RNA-sequencing revealed that DDA/TDB/3M-052 liposomes initiated early transcription of class-switch regulating genes directly in pre-germinal center B cells. Mixed bone marrow chimeras further demonstrated that this adjuvant did not require Th1 cells for IgG2a/c switching, but rather facilitated TLR7-dependent T-bet programming directly in B cells. This study underlines that adjuvant-directed IgG2a/c class-switching in vivo can occur in the absence of T-cell help, via direct activation of TLR7 on B cells and positions DDA/TDB/3M-052 as a powerful adjuvant capable of eliciting type I-like immunity in B cells without strong induction of Th1 responses.
Collapse
Affiliation(s)
- Julie Zimmermann
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | - Katharina Wørzner
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ricki T Krog
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Safia Guleed
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Tu Hu
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Rasmus Mortensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jes Dietrich
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Sara M Ø Solbak
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
10
|
Rios LE, Lokugamage N, Choudhuri S, Chowdhury IH, Garg NJ. Subunit nanovaccine elicited T cell functional activation controls Trypanosoma cruzi mediated maternal and placental tissue damage and improves pregnancy outcomes in mice. NPJ Vaccines 2023; 8:188. [PMID: 38104118 PMCID: PMC10725459 DOI: 10.1038/s41541-023-00782-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
This study investigated a candidate vaccine effect against maternal Trypanosoma cruzi (Tc) infection and improved pregnancy outcomes. For this, TcG2 and TcG4 were cloned in a nanoplasmid optimized for delivery, antigen expression, and regulatory compliance (nano2/4 vaccine). Female C57BL/6 mice were immunized with nano2/4, infected (Tc SylvioX10), and mated 7-days post-infection to enable fetal development during the maternal acute parasitemia phase. Females were euthanized at E12-E17 (gestation) days. Splenic and placental T-cell responses were monitored by flow cytometry. Maternal and placental/fetal tissues were examined for parasites by qPCR and inflammatory infiltrate by histology. Controls included age/immunization-matched non-pregnant females. Nano2/4 exhibited no toxicity and elicited protective IgG2a/IgG1 response in mice. Nano2/4 signaled a splenic expansion of functionally active CD4+ effector/effector memory (Tem) and central memory (Tcm) cells in pregnant mice. Upon challenge infection, nano2/4 increased the splenic CD4+ and CD8+T cells in all mice and increased the proliferation of CD4+Tem, CD4+Tcm, and CD8+Tcm subsets producing IFNγ and cytolytic molecules (PRF1, GZB) in pregnant mice. A balanced serum cytokines/chemokines response and placental immune characteristics indicated that pregnancy prevented the overwhelming damaging immune response in mice. Importantly, pregnancy itself resulted in a significant reduction of parasites in maternal and fetal tissues. Nano2/4 was effective in arresting the Tc-induced tissue inflammatory infiltrate, necrosis, and fibrosis in maternal and placental tissues and improving maternal fertility, placental efficiency, and fetal survival. In conclusion, we show that maternal nano2/4 vaccination is beneficial in controlling the adverse effects of Tc infection on maternal health, fetal survival, and pregnancy outcomes.
Collapse
Affiliation(s)
- Lizette Elaine Rios
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
- Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA
| | - Nandadeva Lokugamage
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Subhadip Choudhuri
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Imran Hussain Chowdhury
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, USA.
- Institute for Human Infections and Immunity (IHII), UTMB, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences (SIVS), UTMB, Galveston, TX, USA.
| |
Collapse
|
11
|
Limnander A, Kaur N, Asrat S, Tasker C, Boyapati A, Ben LH, Janczy J, Pedraza P, Abreu P, Chen WC, Godin S, Daniel BJ, Chin H, DeVeaux M, Rodriguez Lorenc K, Sirulnik A, Harari O, Stahl N, Sleeman MA, Murphy AJ, Yancopoulos GD, Orengo JM. A therapeutic strategy to target distinct sources of IgE and durably reverse allergy. Sci Transl Med 2023; 15:eadf9561. [PMID: 38091405 DOI: 10.1126/scitranslmed.adf9561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Immunoglobulin E (IgE) is a key driver of type 1 hypersensitivity reactions and allergic disorders, which are globally increasing in number and severity. Although eliminating pathogenic IgE may be a powerful way to treat allergy, no therapeutic strategy reported to date can fully ablate IgE production. Interleukin-4 receptor α (IL-4Rα) signaling is required for IgE class switching, and IL-4Rα blockade gradually reduces, but does not eliminate, IgE. The persistence of IgE after IL-4Rα blockade may be due to long-lived IgE+ plasma cells that maintain serological memory to allergens and thus may be susceptible to plasma cell-targeted therapeutics. We demonstrate that transient administration of a B cell maturation antigen x CD3 (BCMAxCD3) bispecific antibody markedly depletes IgE, as well as other immunoglobulins, by ablating long-lived plasma cells, although IgE and other immunoglobulins rapidly rebound after treatment. Concomitant IL-4Rα blockade specifically and durably prevents the reemergence of IgE by blocking IgE class switching while allowing the restoration of other immunoglobulins. Moreover, this combination treatment prevented anaphylaxis in mice. Together with additional cynomolgus monkey and human data, our studies demonstrate that allergic memory is primarily maintained by both non-IgE+ memory B cells that require class switching and long-lived IgE+ plasma cells. Our combination approach to durably eliminate pathogenic IgE has potential to benefit allergy in humans while preserving antibody-mediated immunity.
Collapse
Affiliation(s)
| | - Navneet Kaur
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Carley Tasker
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Anita Boyapati
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Li-Hong Ben
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - John Janczy
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Pablo Abreu
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Wen-Chi Chen
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Stephen Godin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Harvey Chin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Olivier Harari
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Neil Stahl
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| |
Collapse
|
12
|
Gholami M, Sadegh M, Koroush-Arami M, Norouzi S, Arismani RJ, Asadi E, Amini M, Khodayari N. Targeting memory loss with aspirin, a molecular mechanism perspective for future therapeutic approaches. Inflammopharmacology 2023; 31:2827-2842. [PMID: 37924473 DOI: 10.1007/s10787-023-01347-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/19/2023] [Indexed: 11/06/2023]
Abstract
Acetylsalicylic acid (ASA), also known as aspirin, was discovered in 1897 as an acetylated form of salicylate. It has been widely used for its anti-inflammatory and antiplatelet effects. It is commonly used for its cardiovascular benefits and is prescribed as secondary prophylaxis after a heart attack. Furthermore, low-dose, long-term ASA is used to reduce the risk of heart attack and stroke in individuals without prior cardiovascular disease. Acetylsalicylic acid acts as a non-selective inhibitor of cyclooxygenase (COX), which inhibits the synthesis of prostaglandins and prevents pro-inflammatory cytokines. Findings suggest that targeting cytokines and growth factors could be a potential therapeutic strategy for reducing neuroinflammation and slowing down the progression of dementia. Additionally, prostaglandins contribute to synaptic plasticity and can act as retrograde messengers in synapses. Research has implicated COX-1, one of the isoforms of the enzyme, in neuroinflammation and neurodegenerative disorders. The inhibition of COX-1 might potentially prevent impairments in working memory and reduce neuroinflammation caused by beta-amyloid proteins in some conditions, such as Alzheimer's disease (AD). Cyclooxygenase-2, an inducible form of the enzyme, is expressed in cortical and hippocampal neurons and is associated with long-term synaptic plasticity. The inhibition or knockout of COX-2 has been shown to decrease long-term potentiation, a process involved in memory formation. Studies have also demonstrated that the administration of COX-2 inhibitors impairs cognitive function and memory acquisition and recall in animal models. There remains a debate regarding the effects of aspirin on dementia and cognitive decline. Although some studies suggest a possible protective effect of non-steroidal anti-inflammatory drugs, including aspirin, against the development of AD, others have shown inconsistent evidence. This review provides an overview of the effects of ASA or its active metabolite salicylate on learning, memory, and synaptic plasticity.
Collapse
Affiliation(s)
- Masoumeh Gholami
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Mehdi Sadegh
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Masoumeh Koroush-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Rasoul Jafari Arismani
- Department of Urologic Surgery, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Erfan Asadi
- Medical Student, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mohammad Amini
- Medical Student, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nahid Khodayari
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
13
|
Ding Z, Mulder J, Robinson MJ. The origins and longevity of IgE responses as indicated by serological and cellular studies in mice and humans. Allergy 2023; 78:3103-3117. [PMID: 37417548 PMCID: PMC10952832 DOI: 10.1111/all.15799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/02/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023]
Abstract
The existence of long-lived IgE antibody-secreting cells (ASC) is contentious, with the maintenance of sensitization by the continuous differentiation of short-lived IgE+ ASC a possibility. Here, we review the epidemiological profile of IgE production, and give an overview of recent discoveries made on the mechanisms regulating IgE production from mouse models. Together, these data suggest that for most individuals, in most IgE-associated diseases, IgE+ ASC are largely short-lived cells. A subpopulation of IgE+ ASC in humans is likely to survive for tens of months, although due to autonomous IgE B cell receptor (BCR) signaling and antigen-driven IgE+ ASC apoptosis, in general IgE+ ASC probably do not persist for the decades that other ASC are inferred to do. We also report on recently identified memory B cell transcriptional subtypes that are the likely source of IgE in ongoing responses, highlighting the probable importance of IL-4Rα in their regulation. We suggest the field should look at dupilumab and other drugs that prohibit IgE+ ASC production as being effective treatments for IgE-mediated aspects of disease in most individuals.
Collapse
Affiliation(s)
- Zhoujie Ding
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | - Jesse Mulder
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | | |
Collapse
|
14
|
Dasari V, McNeil LK, Beckett K, Solomon M, Ambalathingal G, Thuy TL, Panikkar A, Smith C, Steinbuck MP, Jakubowski A, Seenappa LM, Palmer E, Zhang J, Haqq CM, DeMuth PC, Khanna R. Lymph node targeted multi-epitope subunit vaccine promotes effective immunity to EBV in HLA-expressing mice. Nat Commun 2023; 14:4371. [PMID: 37553346 PMCID: PMC10409721 DOI: 10.1038/s41467-023-39770-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/28/2023] [Indexed: 08/10/2023] Open
Abstract
The recent emergence of a causal link between Epstein-Barr virus (EBV) and multiple sclerosis has generated considerable interest in the development of an effective vaccine against EBV. Here we describe a vaccine formulation based on a lymph node targeting Amphiphile vaccine adjuvant, Amphiphile-CpG, admixed with EBV gp350 glycoprotein and an engineered EBV polyepitope protein that includes 20 CD8+ T cell epitopes from EBV latent and lytic antigens. Potent gp350-specific IgG responses are induced in mice with titers >100,000 in Amphiphile-CpG vaccinated mice. Immunization including Amphiphile-CpG also induces high frequencies of polyfunctional gp350-specific CD4+ T cells and EBV-specific CD8+ T cells that are 2-fold greater than soluble CpG and are maintained for >7 months post immunization. This combination of broad humoral and cellular immunity against multiple viral determinants is likely to provide better protection against primary infection and control of latently infected B cells leading to protection against the development of EBV-associated diseases.
Collapse
Affiliation(s)
- Vijayendra Dasari
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia.
| | | | - Kirrilee Beckett
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - Matthew Solomon
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - George Ambalathingal
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - T Le Thuy
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - Archana Panikkar
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - Caitlyn Smith
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | | | | | - Jeff Zhang
- Elicio Therapeutics, Inc, Boston, MA, USA
| | | | | | - Rajiv Khanna
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia.
| |
Collapse
|
15
|
Mambelli F, Marinho FV, Andrade JM, de Araujo ACVSC, Abuna RPF, Fabri VMR, Santos BPO, da Silva JS, de Magalhães MTQ, Homan EJ, Leite LCC, Dias GB, Heck N, Mendes DAGB, Mansur DS, Báfica A, Oliveira SC. Recombinant Bacillus Calmette-Guérin Expressing SARS-CoV-2 Chimeric Protein Protects K18-hACE2 Mice against Viral Challenge. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1925-1937. [PMID: 37098890 PMCID: PMC10247535 DOI: 10.4049/jimmunol.2200731] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/24/2023] [Indexed: 04/27/2023]
Abstract
COVID-19 has accounted for more than 6 million deaths worldwide. Bacillus Calmette-Guérin (BCG), the existing tuberculosis vaccine, is known to induce heterologous effects over other infections due to trained immunity and has been proposed to be a potential strategy against SARS-CoV-2 infection. In this report, we constructed a recombinant BCG (rBCG) expressing domains of the SARS-CoV-2 nucleocapsid and spike proteins (termed rBCG-ChD6), recognized as major candidates for vaccine development. We investigated whether rBCG-ChD6 immunization followed by a boost with the recombinant nucleocapsid and spike chimera (rChimera), together with alum, provided protection against SARS-CoV-2 infection in K18-hACE2 mice. A single dose of rBCG-ChD6 boosted with rChimera associated with alum elicited the highest anti-Chimera total IgG and IgG2c Ab titers with neutralizing activity against SARS-CoV-2 Wuhan strain when compared with control groups. Importantly, following SARS-CoV-2 challenge, this vaccination regimen induced IFN-γ and IL-6 production in spleen cells and reduced viral load in the lungs. In addition, no viable virus was detected in mice immunized with rBCG-ChD6 boosted with rChimera, which was associated with decreased lung pathology when compared with BCG WT-rChimera/alum or rChimera/alum control groups. Overall, our study demonstrates the potential of a prime-boost immunization system based on an rBCG expressing a chimeric protein derived from SARS-CoV-2 to protect mice against viral challenge.
Collapse
Affiliation(s)
- Fábio Mambelli
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio V. Marinho
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juvana M. Andrade
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana C. V. S. C. de Araujo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo P. F. Abuna
- Platform of Bi-Institutional Research in Translational Medicine, Oswaldo Cruz Foundation-Fiocruz, Ribeirão Preto, São Paulo, Brazil
| | - Victor M. R. Fabri
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno P. O. Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - João S. da Silva
- Platform of Bi-Institutional Research in Translational Medicine, Oswaldo Cruz Foundation-Fiocruz, Ribeirão Preto, São Paulo, Brazil
| | - Mariana T. Q. de Magalhães
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - E. Jane Homan
- ioGenetics LLC, Madison, Wisconsin, United States of America
| | | | - Greicy B.M. Dias
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Nicoli Heck
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniel A. G. B. Mendes
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniel S. Mansur
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - André Báfica
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Sergio C. Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Chen T, Ding Z, Lan J, Wong G. Advances and perspectives in the development of vaccines against highly pathogenic bunyaviruses. Front Cell Infect Microbiol 2023; 13:1174030. [PMID: 37274315 PMCID: PMC10234439 DOI: 10.3389/fcimb.2023.1174030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Increased human activities around the globe and the rapid development of once rural regions have increased the probability of contact between humans and wild animals. A majority of bunyaviruses are of zoonotic origin, and outbreaks may result in the substantial loss of lives, economy contraction, and social instability. Many bunyaviruses require manipulation in the highest levels of biocontainment, such as Biosafety Level 4 (BSL-4) laboratories, and the scarcity of this resource has limited the development speed of vaccines for these pathogens. Meanwhile, new technologies have been created, and used to innovate vaccines, like the mRNA vaccine platform and bioinformatics-based antigen design. Here, we summarize current vaccine developments for three different bunyaviruses requiring work in the highest levels of biocontainment: Crimean-Congo Hemorrhagic Fever Virus (CCHFV), Rift Valley Fever Virus (RVFV), and Hantaan virus (HTNV), and provide perspectives and potential future directions that can be further explored to advance specific vaccines for humans and livestock.
Collapse
Affiliation(s)
- Tong Chen
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhe Ding
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaming Lan
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Gary Wong
- Viral Hemorrhagic Fevers Research Unit, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, China
| |
Collapse
|
17
|
Araujo IL, Piraine REA, Fischer G, Leite FPL. Recombinant BoHV-5 glycoprotein (rgD5) elicits long-lasting protective immunity in cattle. Virology 2023; 584:44-52. [PMID: 37244054 DOI: 10.1016/j.virol.2023.04.004] [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: 07/02/2022] [Revised: 03/27/2023] [Accepted: 04/13/2023] [Indexed: 05/29/2023]
Abstract
BoHV-5 is a worldwide distributed pathogen usually associated with a lethal neurological disease in dairy and beef cattle resulting in important economic losses due to the cattle industry. Using recombinant gD5, we evaluated the long-duration humoral immunity of the recombinant vaccines in a cattle model. Here we report that two doses of intramuscular immunization, particularly with the rgD5ISA vaccine, induce long-lasting antibody responses. Recombinant gD5 antigen elicited tightly mRNA transcription of the Bcl6 and the chemokine receptor CXCR5 which mediate memory B cells and long-lived plasma cells in germinal centers. In addition, using an in-house indirect ELISA we observed higher and earlier responses of rgD5-specific IgG antibody and the upregulation of mRNA transcription of IL2, IL4, IL10, IL15, and IFN-γ in rgD5 vaccinated cattle, indicating a mixed immune response. We further show that rgD5 immunization protects against both BoHV -1 and -5. Our findings indicate that the rgD5-based vaccine represents an effective vaccine strategy to induce an efficient control of herpesviruses.
Collapse
Affiliation(s)
- Itauá L Araujo
- Biotechnology Unit, Technological Development Centre, Federal University of Pelotas, Pelotas, Brazil.
| | - Renan E A Piraine
- Biotechnology Unit, Technological Development Centre, Federal University of Pelotas, Pelotas, Brazil.
| | - Geferson Fischer
- Laboratory of Virology and Immunology, Federal University of Federal de Pelotas, Pelotas, RS, Brazil.
| | - Fábio P L Leite
- Biotechnology Unit, Technological Development Centre, Federal University of Pelotas, Pelotas, Brazil; Laboratory of Virology and Immunology, Federal University of Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
18
|
Liu Y, Li B, Zheng X, Xiong D, Ye X. Cancer Vaccines Based on Fluorine-Modified KH-1 Elicit Robust Immune Response. Molecules 2023; 28:molecules28041934. [PMID: 36838925 PMCID: PMC9963332 DOI: 10.3390/molecules28041934] [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: 01/12/2023] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
KH-1 is a tumor-associated carbohydrate antigen (TACA), which serves as a valuable target of antitumor vaccines for cancer immunotherapies. However, most TACAs are thymus-independent antigens (TD-Ag), and they tend to induce immunological tolerance, leading to their low immunogenicity. To overcome these problems, some fluorinated derivatives of the KH-1 antigen were designed, synthesized, and conjugated to the carrier protein CRM197 to form glycoconjugates, which were used for immunological studies with Freund's adjuvant. The results showed that fluorine-modified N-acyl KH-1 conjugates can induce higher titers of antibodies, especially IgG, which can recognize KH-1-positive cancer cells and can eliminate cancer cells through complement-dependent cytotoxicity (CDC). The trifluoro-modified KH-1-TF-CRM197 showed great potential as an anticancer vaccine candidate.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China
| | - Bohan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China
| | - Xiujing Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China
| | - Decai Xiong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China
- The NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, 27 Shanda Nanlu, Jinan 250100, China
- Correspondence: (D.X.); (X.Y.)
| | - Xinshan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China
- Correspondence: (D.X.); (X.Y.)
| |
Collapse
|
19
|
Reuter S, Raspe J, Uebner H, Contoyannis A, Pastille E, Westendorf AM, Caso GC, Cover TL, Müller A, Taube C. Treatment with Helicobacter pylori-derived VacA attenuates allergic airway disease. Front Immunol 2023; 14:1092801. [PMID: 36761723 PMCID: PMC9902502 DOI: 10.3389/fimmu.2023.1092801] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/03/2023] [Indexed: 01/26/2023] Open
Abstract
Background Asthma is an incurable heterogeneous disease with variations in clinical and underlying immunological phenotype. New approaches could help to support existing therapy concepts. Neonatal infection of mice with Helicobacter pylori or administration of H. pylori-derived extracts or molecules after birth have been shown to prevent the development of allergic airway disease later in life. This study evaluated the potential therapeutic efficacy of H. pylori vacuolating cytotoxin A (VacA) in allergic airway inflammation and investigated the underlying immunological mechanisms for its actions. Methods Murine models of allergic airway diseases, and murine and human in vitro models were used. Results In both an acute model and a therapeutic house dust mite model of allergic airway disease, treatment with H. pylori-derived VacA reduced several asthma hallmarks, including airway hyperresponsiveness, inflammation and goblet cell metaplasia. Flow cytometry and ELISA analyses revealed induction of tolerogenic dendritic cells (DC) and FoxP3 positive regulatory T cells (Tregs), and a shift in the composition of allergen-specific immunoglobulins. Depletion of Tregs during treatment with VacA reversed treatment-mediated suppression of allergic airway disease. Human monocyte derived DCs (moDC) that were exposed to VacA induced Tregs in co-cultured naïve autologous T cells, replicating key observations made in vivo. Conclusion H. pylori-derived VacA suppressed allergic airway inflammation via induction of Tregs in both allergic airway disease models. These data suggest that the immunomodulatory activity of VacA could potentially be exploited for the prevention and treatment of allergic airway disease.
Collapse
Affiliation(s)
- Sebastian Reuter
- Department of Pulmonary Medicine, Experimental Pneumology, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Jonas Raspe
- Department of Pulmonary Medicine, Experimental Pneumology, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Hendrik Uebner
- Department of Pulmonary Medicine, Experimental Pneumology, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Alexandros Contoyannis
- Department of Pulmonary Medicine, Experimental Pneumology, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Eva Pastille
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Astrid M. Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Georgia C. Caso
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Timothy L. Cover
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Affairs Tennessee Valley Healthcare System Nashville, Nashville, TN, United States
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Christian Taube
- Department of Pulmonary Medicine, Experimental Pneumology, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| |
Collapse
|
20
|
Lamichhane P, Schmidt ME, Terhüja M, Varga SM, Snider TA, Rostad CA, Oomens AGP. A live single-cycle RSV vaccine expressing prefusion F protein. Virology 2022; 577:51-64. [PMID: 36306605 PMCID: PMC10104964 DOI: 10.1016/j.virol.2022.10.003] [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: 06/07/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Live-attenuated Respiratory syncytial virus (RSV) vaccines given intranasally have potential to provide comprehensive protection, including lung-resident immunity. It has however proven challenging to impart both sufficient safety and efficacy in a vaccine. To achieve the latter, we used a trans-complementing approach to generate live single-cycle RSV vaccines expressing the prefusion form (preF) of the viral fusion protein (F), either membrane-anchored or secreted. Both viruses were tested for their ability to induce a protective immune response in mice after intranasal prime-boost vaccination. The secreted preF vaccine failed to induce a protective response. The anchored preF vaccine induced anti-preF antibodies and antiviral T cells, and protected mice from lung pathology and viral shedding after challenge. Neither vaccine induced anti-G antibodies, for reasons unknown. In spite of the latter and single-cycle replication, the membrane-anchored preF vaccine was protective and demonstrates potential for development of an efficacious live vaccine with a stable safety phenotype.
Collapse
Affiliation(s)
- Pramila Lamichhane
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, 52242, USA
| | - Megolhubino Terhüja
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, 52242, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, 52242, USA; Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Timothy A Snider
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Christina A Rostad
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA, 30329, USA
| | - Antonius G P Oomens
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
21
|
Guzelj S, Weiss M, Slütter B, Frkanec R, Jakopin Ž. Covalently Conjugated NOD2/TLR7 Agonists Are Potent and Versatile Immune Potentiators. J Med Chem 2022; 65:15085-15101. [DOI: 10.1021/acs.jmedchem.2c00808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Samo Guzelj
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Matjaž Weiss
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Ruža Frkanec
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
22
|
Ortega-Rivera OA, Shin MD, Moreno-Gonzalez MA, Pokorski JK, Steinmetz NF. A single-dose Qβ VLP vaccine against S100A9 protein reduces atherosclerosis in a preclinical model. ADVANCED THERAPEUTICS 2022; 5:2200092. [PMID: 36570039 PMCID: PMC9783282 DOI: 10.1002/adtp.202200092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Indexed: 12/30/2022]
Abstract
The standard therapy for cardiovascular disease (CVD) is the administration of statins to reduce plasma cholesterol levels, but this requires lifelong treatment. We developed a CVD vaccine candidate that targets the pro-inflammatory mediator calprotectin by eliciting antibodies against the S100A9 protein. The vaccine, based on bacteriophage Qβ virus-like particles (VLPs) displaying S100A9 peptide epitopes, was formulated as a slow-release PLGA:VLP implant by hot-melt extrusion. The single-dose implant elicited S100A9-specific antibody titers comparable to a three-dose injection schedule with soluble VLPs. In an animal model of CVD (ApoE-/- mice fed on a high-fat diet), the implant reduced serum levels of calprotectin, IL-1β, IL-6 and MCP-1, resulting in less severe aortic lesions. This novel implant was therefore able to attenuate atherosclerosis over a sustained period and offers a novel and promising strategy to replace the repetitive administration of statins for the treatment of CVD.
Collapse
Affiliation(s)
- Oscar A. Ortega-Rivera
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
| | - Matthew D. Shin
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
| | - Miguel A. Moreno-Gonzalez
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
| | - Jonathan K. Pokorski
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Institute for Materials Discovery and Design, University of California-San Diego, La Jolla CA 92039, USA
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Institute for Materials Discovery and Design, University of California-San Diego, La Jolla CA 92039, USA
- Department of Bioengineering, University of California-San Diego, La Jolla CA 92039, USA
- Department of Radiology, University of California-San Diego, La Jolla CA 92039, USA
- Moores Cancer Center, University of California-San Diego, La Jolla CA 92039, USA
| |
Collapse
|
23
|
Tambunlertchai S, Geary SM, Salem AK. Topically Applied Resiquimod versus Imiquimod as a Potential Adjuvant in Melanoma Treatment. Pharmaceutics 2022; 14:pharmaceutics14102076. [PMID: 36297510 PMCID: PMC9611754 DOI: 10.3390/pharmaceutics14102076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 01/28/2023] Open
Abstract
Melanoma is the most lethal form of skin cancer and surgery remains the preferred and most effective treatment. Nevertheless, there are cases where surgery is not a viable method and alternative treatments are therefore adopted. One such treatment that has been tested is topical 5% imiquimod (IMQ) cream, which, although showing promise as a treatment for melanoma, has been found to have undesirable off-target effects. Resiquimod (RSQ) is an immunomodulatory molecule that can activate immune responses by binding to Toll-like receptors (TLR) 7 and 8 and may be more effective than IMQ in the context of melanoma treatment. RSQ can cross the stratum corneum (SC) easily without requiring pretreatment of the skin. In a gel formulation, RSQ has been studied as a monotherapy and adjuvant for melanoma treatment in pre-clinical studies and as an adjuvant in clinical settings. Although side effects of RSQ in gel formulation were also reported, they were never severe enough for the treatment to be suspended. In this review, we discuss the potential use of RSQ as an adjuvant for melanoma treatment.
Collapse
|
24
|
Shen Y, Zhang N, Yang Y, Hong S, Bachert C. Local Immunoglobulin E in nasal polyps: Role and modulation. Front Immunol 2022; 13:961503. [PMID: 36159836 PMCID: PMC9492990 DOI: 10.3389/fimmu.2022.961503] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
In the airway, IgE is traditionally regarded as a key mediator in allergic diseases, such as AR and allergic asthma. However, growing evidence demonstrates the importance of local IgE in airway inflammatory diseases, irrespective of the presence of allergy. In this review, we discuss the most recent evidence for IgE in chronic rhinosinusitis with nasal polyps(CRSwNP), including the local IgE’s characteristics, the modulation of its synthesis, and its function. The levels of local IgE are significantly elevated in polyps independently of IgE serum levels and atopic status. Local IgE, which is correlated with type 2 inflammation, is polyclonal and functional. IgE is produced by active B cells and is dependent on the class switch recombination(CSR). In NPs, this process is triggered by not only allergens but also microbial colonization, especially the superantigen- Staphylococcus aureus. The production of local IgE is modulated by lymphocytes(such as Tfh, ILC2s, iTreg), cytokines(such as IL-4, IL-13, IFN-γ, TGF-β, IL-2, IL-21), transcription factors, and B cell-intrinsic factor. Due to the central role of IgE in NPs, it is regarded as an ideal target for therapy and has been proved to be clinically successful. Based on this knowledge, we believe that exploring the trigger and regulatory factors for the activation of local B cells and CSR to IgE will provide more valuable information for us to recognize the pathological mechanisms of local IgE and offer the possible option for new therapeutic targets of nasal polyps.
Collapse
Affiliation(s)
- Yang Shen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Zhang
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
| | - Yucheng Yang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Suling Hong
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Claus Bachert
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
- Division of Otorhinolaryngology Diseases, Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
- *Correspondence: Claus Bachert,
| |
Collapse
|
25
|
Japanese Encephalitis DNA Vaccines with Epitope Modification Reduce the Induction of Cross-Reactive Antibodies against Dengue Virus and Antibody-Dependent Enhancement of Dengue Virus Infection. Vaccines (Basel) 2022; 10:vaccines10091411. [PMID: 36146489 PMCID: PMC9506301 DOI: 10.3390/vaccines10091411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/08/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
Infection with viruses belonging to the genus Flavivirus, such as Japanese encephalitis virus (JEV) and dengue virus (DENV), is a worldwide health problem. Vaccines against JEV and DENV are currently available. However, the dengue vaccine possibly increases the risk of severe dengue due to antibody-dependent enhancement (ADE). Moreover, the Japanese encephalitis (JE) vaccine reportedly induces cross-reactive ADE-prone antibodies against DENV, potentially leading to symptomatic dengue. Therefore, it is necessary to eliminate the risk of ADE through vaccination. In this study, we attempted to develop a JE vaccine that does not induce ADE of DENV infection using an epitope modification strategy. We found that an ADE-prone monoclonal antibody cross-reactive to DENV and JEV recognizes the 106th amino acid residue of the E protein of JEV (E-106). The JE DNA vaccine with a mutation at E-106 (E-106 vaccine) induced comparable neutralizing antibody titers against JEV to those induced by the wild-type JE DNA vaccine. Meanwhile, the E-106 vaccine induced 64-fold less cross-reactive ADE-prone antibodies against DENV. The mutation did not compromise the protective efficacy of the vaccine in the lethal JEV challenge experiment. Altogether, the modification of a single amino acid residue identified in this study helped in the development of an ADE-free JE vaccine.
Collapse
|
26
|
van Haren SD, Pedersen GK, Kumar A, Ruckwardt TJ, Moin S, Moore IN, Minai M, Liu M, Pak J, Borriello F, Doss-Gollin S, Beijnen EMS, Ahmed S, Helmel M, Andersen P, Graham BS, Steen H, Christensen D, Levy O. CAF08 adjuvant enables single dose protection against respiratory syncytial virus infection in murine newborns. Nat Commun 2022; 13:4234. [PMID: 35918315 PMCID: PMC9346114 DOI: 10.1038/s41467-022-31709-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/30/2022] [Indexed: 11/09/2022] Open
Abstract
Respiratory syncytial virus is a leading cause of morbidity and mortality in children, due in part to their distinct immune system, characterized by impaired induction of Th 1 immunity. Here we show application of cationic adjuvant formulation CAF08, a liposomal vaccine formulation tailored to induce Th 1 immunity in early life via synergistic engagement of Toll-like Receptor 7/8 and the C-type lectin receptor Mincle. We apply quantitative phosphoproteomics to human dendritic cells and reveal a role for Protein Kinase C-δ for enhanced Th1 cytokine production in neonatal dendritic cells and identify signaling events resulting in antigen cross-presentation. In a murine in vivo model a single immunization at birth with CAF08-adjuvanted RSV pre-fusion antigen protects newborn mice from RSV infection by induction of antigen-specific CD8+ T-cells and Th1 cells. Overall, we describe a pediatric adjuvant formulation and characterize its mechanism of action providing a promising avenue for development of early life vaccines against RSV and other respiratory viral pathogens.
Collapse
Affiliation(s)
- Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Azad Kumar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Syed Moin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mark Liu
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Jensen Pak
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- Generate Biomedicines, Cambridge, MA, USA
| | - Simon Doss-Gollin
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Elisabeth M S Beijnen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Saima Ahmed
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michaela Helmel
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Andersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanno Steen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| |
Collapse
|
27
|
Augusto Sanches Roque G, Esteves Zorgi N, Janaína Soares Rocha F, Flóro E Silva M, Fernanda Araújo T, Ruiz Abánades D, Giorgio S. Evaluation of prime and prime-boost immunization strategies in BALB/c mice inoculated with Leishmania infantum transfected with toxic plasmids. Vaccine 2022; 40:4105-4115. [PMID: 35660330 DOI: 10.1016/j.vaccine.2022.05.063] [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: 02/09/2022] [Revised: 04/28/2022] [Accepted: 05/24/2022] [Indexed: 11/29/2022]
Abstract
The etiologic agents of visceral leishmaniasis are Leishmania infantum and Leishmania donovani. Despite the variety of drugs available to treat leishmaniasis, most lead to serious adverse effects, and resistance to these drugs has been reported. Currently, no leishmaniasis vaccine is available for humans. That is why the group developed transgenic L. infantum promastigote lines, which express toxic proteins after differentiation into amastigotes. That is why group developed the pFL-AMA plasmid and transfected it into L. Infantum promastigotes. This plasmid was expressed only in the amastigote form of the parasite. Sequences encoding toxic proteins (active bovine trypsin and egg avidin) were inserted in this plasmid, and the transfected parasites died after the differentiation process. In this study, two immunization protocols were performed in BALB/c mice: prime and prime-boost immunization prior to challenge with the wild-type L. infantum (WT). The parasite burdens in the spleen, liver, and bone marrow were evaluated to verify immunological protection. Histopathological analysis of the spleen and liver and the humoral immune response were also performed. The data showed that the parasite burden was reduced in prime-boosted mice in the spleen, liver, and bone marrow, indicating that mice immunized with two doses of the transfected parasites were satisfactorily protected. High levels of IgG, IgG1, and IgG2a antibodies were observed, as well as the presence of anti-inflammatory cytokine Interleukine-10 and pro-inflammatory cytokine Tumor Necrosis Factor-α (TNF-α) and Interferon-γ (IFN - γ) suggesting a Th1/Th2 mix response, in addition to the presence of multinucleated giant cells in the spleen and lymphocyte infiltration in the liver. Therefore, L. infantum transfected with a toxic plasmid is an excellent vaccine candidate against visceral leishmaniasis and the application of a boost before the challenge promotes greater protection against WT L. infantum infection.
Collapse
Affiliation(s)
| | - Nahiara Esteves Zorgi
- Animal Biology Department, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | | | - Marina Flóro E Silva
- Animal Biology Department, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Thalita Fernanda Araújo
- Animal Biology Department, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Daniel Ruiz Abánades
- Animal Biology Department, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Selma Giorgio
- Animal Biology Department, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
28
|
Zhang Y, Tan W, Lou Z, Huang B, Zhou W, Zhao Y, Zhang J, Liang H, Li N, Zhu X, Ding L, Guo Y, He Z, He Y, Wang Z, Ma B, Ma M, Zhao S, Chang Z, Zhao X, Zheng X, Wu G, Wang H, Yang X. Immunogenicity Evaluating of the Multivalent COVID-19 Inactivated Vaccine against the SARS-CoV-2 Variants. Vaccines (Basel) 2022; 10:vaccines10060956. [PMID: 35746564 PMCID: PMC9228943 DOI: 10.3390/vaccines10060956] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 12/15/2022] Open
Abstract
It has been reported that the novel coronavirus (COVID-19) has caused more than 286 million cases and 5.4 million deaths to date. Several strategies have been implemented globally, such as social distancing and the development of the vaccines. Several severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have appeared, such as Alpha, Beta, Gamma, Delta, and Omicron. With the rapid spread of the novel coronavirus and the rapidly changing mutants, the development of a broad-spectrum multivalent vaccine is considered to be the most effective way to defend against the constantly mutating virus. Here, we evaluated the immunogenicity of the multivalent COVID-19 inactivated vaccine. Mice were immunized by multivalent COVID-19 inactivated vaccine, and the neutralizing antibodies in serum were analyzed. The results show that HB02 + Delta + Omicron trivalent vaccine could provide broad spectrum protection against HB02, Beta, Delta, and Omicron virus. Additionally, the different multivalent COVID-19 inactivated vaccines could enhance cellular immunity. Together, our findings suggest that the multivalent COVID-19 inactivated vaccine can provide broad spectrum protection against HB02 and other virus variants in humoral and cellular immunity, providing new ideas for the development of a broad-spectrum COVID-19 vaccine.
Collapse
Affiliation(s)
- Yuntao Zhang
- China National Biotec Group Company Limited, Beijing 100024, China;
| | - Wenjie Tan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
| | - Zhiyong Lou
- MOE Key Laboratory of Protein Science & Collaborative Innovation Center of Biotherapy, School of Medicine, Tsinghua University, Beijing 100084, China;
| | - Baoying Huang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
| | - Weimin Zhou
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
| | - Yuxiu Zhao
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Jin Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Hongyang Liang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Na Li
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Xiujuan Zhu
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Ling Ding
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Yancen Guo
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Zhenyu He
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Yao He
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Zhanhui Wang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Bo Ma
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Meng Ma
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Suhua Zhao
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Zhen Chang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Xue Zhao
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Guizhen Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
- Correspondence: (G.W.); (H.W.); (X.Y.)
| | - Hui Wang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
- Correspondence: (G.W.); (H.W.); (X.Y.)
| | - Xiaoming Yang
- China National Biotec Group Company Limited, Beijing 100024, China;
- Correspondence: (G.W.); (H.W.); (X.Y.)
| |
Collapse
|
29
|
Zagorski K, Chailyan G, Hovakimyan A, Antonyan T, Kiani Shabestari S, Petrushina I, Davtyan H, Cribbs DH, Blurton-Jones M, Masliah E, Agadjanyan MG, Ghochikyan A. Immunogenicity of MultiTEP-Platform-Based Recombinant Protein Vaccine, PV-1950R, Targeting Three B-Cell Antigenic Determinants of Pathological α-Synuclein. Int J Mol Sci 2022; 23:6080. [PMID: 35682759 PMCID: PMC9181659 DOI: 10.3390/ijms23116080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are characterized by the aberrant accumulation of intracytoplasmic misfolded and aggregated α-synuclein (α-Syn), resulting in neurodegeneration associated with inflammation. The propagation of α-Syn aggregates from cell to cell is implicated in the spreading of pathological α-Syn in the brain and disease progression. We and others demonstrated that antibodies generated after active and passive vaccinations could inhibit the propagation of pathological α-Syn in the extracellular space and prevent/inhibit disease/s in the relevant animal models. We recently tested the immunogenicity and efficacy of four DNA vaccines on the basis of the universal MultiTEP platform technology in the DLB/PD mouse model. The antibodies generated by these vaccines efficiently reduced/inhibited the accumulation of pathological α-Syn in the different brain regions and improved the motor deficit of immunized female mice. The most immunogenic and preclinically effective vaccine, PV-1950D, targeting three B-cell epitopes of pathological α-Syn simultaneously, has been selected for future IND-enabling studies. However, to ensure therapeutically potent concentrations of α-Syn antibodies in the periphery of the vaccinated elderly, we developed a recombinant protein-based MultiTEP vaccine, PV-1950R/A, and tested its immunogenicity in young and aged D-line mice. Antibody responses induced by immunizations with the PV-1950R/A vaccine and its homologous DNA counterpart, PV-1950D, in a mouse model of PD/DLB have been compared.
Collapse
Affiliation(s)
- Karen Zagorski
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Gor Chailyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Tatevik Antonyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Sepideh Kiani Shabestari
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
| | - Hayk Davtyan
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - David H. Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; (S.K.S.); (I.P.); (H.D.); (D.H.C.); (M.B.-J.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD 20814, USA;
| | - Michael G. Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; (K.Z.); (G.C.); (A.H.); (T.A.)
| |
Collapse
|
30
|
Gao X, Song Y, Wu J, Lu S, Min X, Liu L, Hu L, Zheng M, Du P, Yu Y, Long H, Wu H, Jia S, Yu D, Lu Q, Zhao M. Iron-dependent epigenetic modulation promotes pathogenic T cell differentiation in lupus. J Clin Invest 2022; 132:152345. [PMID: 35499082 PMCID: PMC9057600 DOI: 10.1172/jci152345] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 03/01/2022] [Indexed: 12/20/2022] Open
Affiliation(s)
- Xiaofei Gao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Yang Song
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Jiali Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Shuang Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Xiaoli Min
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Limin Liu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Longyuan Hu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Meiling Zheng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Pei Du
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Yaqin Yu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Hai Long
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Sujie Jia
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha, China
| | - Di Yu
- Diamantina Institute, The University of Queensland, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| |
Collapse
|
31
|
Liang Z, Luo Z, Chen J, Li B, Li L, Shen C. Bavachin inhibits IL-4 expression by downregulating STAT6 phosphorylation and GATA-3 expression and ameliorates asthma inflammation in an animal model. Immunobiology 2022; 227:152182. [DOI: 10.1016/j.imbio.2022.152182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 11/05/2022]
|
32
|
Krempski JW, Lama JK, Iijima K, Kobayashi T, Matsunaga M, Kita H. A Mouse Model of the “LEAP” Study Reveals a Role for CTLA-4 in Preventing Peanut Allergy Induced by Environmental Peanut Exposure. J Allergy Clin Immunol 2022; 150:425-439.e3. [PMID: 35288169 PMCID: PMC9378358 DOI: 10.1016/j.jaci.2022.02.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/10/2022] [Accepted: 02/03/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND A human study, Learning Early About Peanut Allergy (LEAP), showed that early introduction of peanut products decreases the prevalence of peanut allergy among children. However, the immunologic mechanisms mediating the protective effects of consuming peanut products are not well understood. OBJECTIVE The objective was to develop a mouse model that simulates the LEAP study and investigate the underlying mechanisms for the study observations. METHODS Adult naive BALB/c mice were fed a commercial peanut butter product (Skippy) or buffer control and concomitantly exposed to peanut flour through the airway or skin to mimic environmental exposure. The animals were analyzed for anaphylactic reaction and by molecular and immunologic approaches. RESULTS After exposure to peanut flour through the airway or skin, naive mice developed peanut allergy, as demonstrated by acute and systemic anaphylaxis in response to challenge with peanut extract. Ingestion of Skippy, however, nearly abolished the increase in peanut-specific IgE and IgG and protected animals from developing anaphylaxis. Skippy-fed mice showed reduced numbers of T follicular helper (Tfh) cells and germinal center B cells in their draining lymph nodes, and single-cell RNA sequencing revealed a CD4+ T-cell population expressing cytotoxic T lymphocyte-associated protein 4 (CTLA-4) in these animals. Critically, blocking CTLA-4 with antibody increased levels of peanut-specific antibodies and reversed the protective effects of Skippy. CONCLUSION Ingestion of a peanut product protects mice from peanut allergy induced by environmental exposure to peanuts, and the CTLA-4 pathway, which regulates Tfh cell responses, likely plays a pivotal role in this protection.
Collapse
|
33
|
Takahara T, Tsuyuki T, Satou A, Wada E, Sakurai K, Ueda R, Tsuzuki T. TGFB1 mRNA expression is associated with poor prognosis and specific features of inflammation in ccRCC. Virchows Arch 2022; 480:635-643. [PMID: 35112134 DOI: 10.1007/s00428-021-03256-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 11/12/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022]
Abstract
To determine whether TGFB1 affects the immune microenvironment of ccRCC, we investigated the association between TGFB1 expression and clinicopathological features. Tissue microarray was generated from 158 total or partial nephrectomy samples and 12 tumor-adjacent normal kidney tissue. TGFB1 expression was assessed by RNA in situ hybridization and quantified using ImageJ software. TGFB1 was significantly upregulated in ccRCC tissue than in normal kidney tissues (P = 1.03 × 10-9). Tumors with a high WHO/ISUP grade had higher TGFB1 expression levels (P = 7.05 × 10-3). Of 139 patients with localized ccRCC and whose follow-up data were available, those in the TGFB1-high group displayed significantly shorter relapse-free survival than those in the TGFB1-low group (P = 0.0251). TGFB1 expression was significantly upregulated in patients who developed distant metastasis after surgery (n = 12) than in patients without metastasis (n = 127; P = 0.00167). TGFB1 expression positively correlated with the number of PD-L1-positive cells in the tumor stroma (P = 0.0206, ρ = 0.163). Furthermore, TGFB1 expression was associated with the formation of tertiary lymphoid structures. TGF-β1 is a prognostic indicator of worse outcome for ccRCC and might be a therapeutic target in advanced ccRCC. Our data provide new insights into the association between tumor biology and tumor microenvironment in ccRCC.
Collapse
Affiliation(s)
- Taishi Takahara
- Department of Surgical Pathology, Aichi Medical University Hospital, 1-1, Yazakokarimata,, Nagakute, 480-1195, Japan.
| | - Takuji Tsuyuki
- Department of Surgical Pathology, Aichi Medical University Hospital, 1-1, Yazakokarimata,, Nagakute, 480-1195, Japan
| | - Akira Satou
- Department of Surgical Pathology, Aichi Medical University Hospital, 1-1, Yazakokarimata,, Nagakute, 480-1195, Japan
| | - Eriko Wada
- Department of Surgical Pathology, Aichi Medical University Hospital, 1-1, Yazakokarimata,, Nagakute, 480-1195, Japan
| | - Kaneko Sakurai
- Department of Surgical Pathology, Aichi Medical University Hospital, 1-1, Yazakokarimata,, Nagakute, 480-1195, Japan
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University, Nagakute, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital, 1-1, Yazakokarimata,, Nagakute, 480-1195, Japan
| |
Collapse
|
34
|
Chen CY, Lin YW, Wang SW, Lin YC, Cheng YY, Ren CT, Wong CH, Wu CY. Synthesis of Azido-Globo H Analogs for Immunogenicity Evaluation. ACS CENTRAL SCIENCE 2022; 8:77-85. [PMID: 35106375 PMCID: PMC8796297 DOI: 10.1021/acscentsci.1c01277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Indexed: 06/14/2023]
Abstract
Globo H (GH) is a tumor-associated carbohydrate antigen (TACA), and GH conjugations have been evaluated as potential cancer vaccines. However, like all carbohydrate-based vaccines, low immunogenicity is a major issue. Modifications of the TACA increase its immunogenicity, but the systemic modification on GH is challenging and the synthesis is cumbersome. In this study, we synthesized several azido-GH analogs for evaluation, using galactose oxidase to selectively oxidize C6-OH of the terminal galactose or N-acetylgalactosamine on lactose, Gb3, Gb4, and SSEA3 into C6 aldehyde, which was then transformed chemically to the azido group. The azido-derivatives were further glycosylated to azido-GH analogs by glycosyltransferases coupled with sugar nucleotide regeneration. These azido-GH analogs and native GH were conjugated to diphtheria toxoid cross-reactive material CRM197 for vaccination with C34 adjuvant in mice. Glycan array analysis of antisera indicated that the azido-GH glycoconjugate with azide at Gal-C6 of Lac (1-CRM197) elicited the highest antibody response not only to GH, SSEA3, and SSEA4, which share the common SSEA3 epitope, but also to MCF-7 cancer cells, which express these Globo-series glycans.
Collapse
Affiliation(s)
- Chiang-Yun Chen
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
- Chemical
Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan
- Department
of Chemistry, National Taiwan University, Taipei, 106, Taiwan
| | - Yu-Wei Lin
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Szu-Wen Wang
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 106, Taiwan
| | - Yung-Chu Lin
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Yang-Yu Cheng
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
- Institute
of Biochemistry and Molecular Biology, National
Yang-Ming University, Taipei 112, Taiwan
| | - Chien-Tai Ren
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chi-Huey Wong
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
- Chemical
Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 106, Taiwan
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Chung-Yi Wu
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
- Chemical
Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
35
|
Allen CDC. Features of B Cell Responses Relevant to Allergic Disease. THE JOURNAL OF IMMUNOLOGY 2022; 208:257-266. [PMID: 35017215 PMCID: PMC10054180 DOI: 10.4049/jimmunol.2100988] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/04/2021] [Indexed: 01/16/2023]
Abstract
This Brief Review delves into B cell responses in the context of allergy. The primary contribution of B cells to allergy is the production of IgE, the Ab isotype that triggers immediate hypersensitivity reactions through the release of mediators from mast cells and basophils. B cells may also have protective roles in allergy, such as through the production of IgG or as regulatory B cells. In this review, I focus on the basic principles of B cell differentiation and discuss features relevant to allergic immune responses. In particular, I discuss: (1) class-switch recombination; (2) plasma cell differentiation; (3) germinal centers and affinity maturation; and (4) memory B cells and recall responses, with an emphasis on IgE, IgG1, and IgG4. I also consider how B cells may contribute to allergic responses independent of Ab production-for example, by serving as APCs.
Collapse
Affiliation(s)
- Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA; and Department of Anatomy, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
36
|
Borchard JL, Conrad NL, Pinto NB, Moura MQD, Berne MEA, Leite FPL. Acute and chronic immunomodulatory response mechanisms against Toxocara canis larvae infection in mice. REVISTA BRASILEIRA DE PARASITOLOGIA VETERINÁRIA 2022. [DOI: 10.1590/s1984-29612022056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract The objective of this work was to evaluate the early and late immunological modulation of an experimental infection of T. canis larvae in mice. Mice were infected with 100 infective larvae and euthanized at different period: 24, 48 hours post infection (HPI), 15- and 30 days post infection (DPI). The humoral response was evaluated by indirect ELISA. Quantitative RT–PCR (qPCR) was used to quantify the mRNA transcription of cytokines IL4, IL10, IL12 and Ym1 in the early and late infection periods. Infection with T. canis was able to generate specific total IgG at 15- and 30- DPI. Analyzing the IgG isotype revealed a significant differentiation for IgG1 compared with IgG2a, IgG2b and IgG3, characterizing a Th-2 response. Evaluating the gene transcription at the early phase of infection, higher transcription levels of IL10, IL4 and Ym1 and a downregulation of IL12 were observed. By the late phase, increased transcription levels of IL4, Ym1 and IL12 were observed, and downregulation of IL-10 transcription was observed. The data obtained suggest that during experimental infection with T. canis, the participation of the IL4, IL10, IL12 cytokines and Ym1 can play an important role in T. canis immunomodulation.
Collapse
|
37
|
Park JE, Shin HJ. Immunogenicity of replication-deficient vesicular stomatitis virus based rabies vaccine in mice. Vet Q 2021; 41:202-209. [PMID: 33985418 PMCID: PMC8172215 DOI: 10.1080/01652176.2021.1930277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/02/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Rabies is a viral disease that causes severe neurological manifestations both in humans and various mammals. Although inactivated and/or attenuated vaccines have been developed and widely used around the world, there are still concerns with regard to their safety, efficacy, and costs. OBJECTIVE As demand has grown for a new rabies vaccine, we have developed a new vesicular stomatitis viruses (VSVs) based rabies vaccine that replaces glycoproteins with rabies virus (RABV) glycoprotein (GP), or so-called VSV/RABV-GP. METHODS VSV/RABV-GP production was measured by sandwich ELISA. The generation of VSV/RABV-GP was evaluated with GP-specific antibodies and reduced transduction with GP-specific neutralizing antibodies. Virus entry was quantified by measuring the luciferase levels at 18-h post-transduction. BALB/c mice (three groups of six mice each) were intraperitoneally immunized with PBS, RABA, or VSV/RABV-GP at 0 and 14 days. At 28 days post-immunization serology was performed. Statistical significance was calculated using the Holm-Sidak multiple Student's t test. RESULTS Mice immunized with VSV/RABV-GP produced IgM and IgG antibodies, whereas IgM titers were significantly higher in mice immunized with VSV/RABV-GP compared to inactivated RABV. The secretion profiles of IgG1 and IgG2a production suggested that VSV/RAVB-GP induces the T helper cell type-2 immune bias. In addition, the average (±SD; n = 3) serum neutralization titers of the inactivated RABV and VSV/RABV-GP groups were 241 ± 40 and 103 ± 54 IU/mL, respectively. CONCLUSION Our results confirm that VSV/RABV-GP could be a new potential vaccination platform for RABV.
Collapse
Affiliation(s)
- Jung-Eun Park
- Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun-Jin Shin
- Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
38
|
Nogueira DS, de Oliveira LM, Amorim CCO, Gazzinelli-Guimarães AC, Barbosa FS, Oliveira FMS, Kraemer L, Mattos M, Cardoso MS, Resende NM, Clímaco MDC, Negrão-Corrêa DA, Faria AMC, Caliari MV, Bueno LL, Gaze S, Russo RC, Gazzinelli-Guimarães PH, Fujiwara RT. Eosinophils mediate SIgA production triggered by TLR2 and TLR4 to control Ascaris suum infection in mice. PLoS Pathog 2021; 17:e1010067. [PMID: 34784389 PMCID: PMC8631680 DOI: 10.1371/journal.ppat.1010067] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 11/30/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
Human ascariasis is the most prevalent but neglected tropical disease in the world, affecting approximately 450 million people. The initial phase of Ascaris infection is marked by larval migration from the host's organs, causing mechanical injuries followed by an intense local inflammatory response, which is characterized mainly by neutrophil and eosinophil infiltration, especially in the lungs. During the pulmonary phase, the lesions induced by larval migration and excessive immune responses contribute to tissue remodeling marked by fibrosis and lung dysfunction. In this study, we investigated the relationship between SIgA levels and eosinophils. We found that TLR2 and TLR4 signaling induces eosinophils and promotes SIgA production during Ascaris suum infection. Therefore, control of parasite burden during the pulmonary phase of ascariasis involves eosinophil influx and subsequent promotion of SIgA levels. In addition, we also demonstrate that eosinophils also participate in the process of tissue remodeling after lung injury caused by larval migration, contributing to pulmonary fibrosis and dysfunction in re-infected mice. In conclusion, we postulate that eosinophils play a central role in mediating host innate and humoral immune responses by controlling parasite burden, tissue inflammation, and remodeling during Ascaris suum infection. Furthermore, we suggest that the use of probiotics can induce eosinophilia and SIgA production and contribute to controlling parasite burden and morbidity of helminthic diseases with pulmonary cycles.
Collapse
Affiliation(s)
- Denise Silva Nogueira
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana Maria de Oliveira
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Centro de Ciências Biológicas e da Saúde, Universidade Federal de Sergipe, Aracajú, Brazil
| | - Chiara Cássia Oliveira Amorim
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Clara Gazzinelli-Guimarães
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Fabrício Marcus Silva Oliveira
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Kraemer
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus Mattos
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mariana Santos Cardoso
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Marianna de Carvalho Clímaco
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ana Maria Caetano Faria
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Vidigal Caliari
- Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Soraya Gaze
- René Rachou Institute, Oswaldo Cruz Foundation–FIOCRUZ, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ricardo Toshio Fujiwara
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
39
|
A live measles-vectored COVID-19 vaccine induces strong immunity and protection from SARS-CoV-2 challenge in mice and hamsters. Nat Commun 2021; 12:6277. [PMID: 34725327 PMCID: PMC8560864 DOI: 10.1038/s41467-021-26506-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 10/05/2021] [Indexed: 11/08/2022] Open
Abstract
Several COVID-19 vaccines have now been deployed to tackle the SARS-CoV-2 pandemic, most of them based on messenger RNA or adenovirus vectors.The duration of protection afforded by these vaccines is unknown, as well as their capacity to protect from emerging new variants. To provide sufficient coverage for the world population, additional strategies need to be tested. The live pediatric measles vaccine (MV) is an attractive approach, given its extensive safety and efficacy history, along with its established large-scale manufacturing capacity. We develop an MV-based SARS-CoV-2 vaccine expressing the prefusion-stabilized, membrane-anchored full-length S antigen, which proves to be efficient at eliciting strong Th1-dominant T-cell responses and high neutralizing antibody titers. In both mouse and golden Syrian hamster models, these responses protect the animals from intranasal infectious challenge. Additionally, the elicited antibodies efficiently neutralize in vitro the three currently circulating variants of SARS-CoV-2.
Collapse
|
40
|
Wigton EJ, Mikami Y, McMonigle RJ, Castellanos CA, Wade-Vallance AK, Zhou SK, Kageyama R, Litterman A, Roy S, Kitamura D, Dykhuizen EC, Allen CD, Hu H, O’Shea JJ, Ansel KM. MicroRNA-directed pathway discovery elucidates an miR-221/222-mediated regulatory circuit in class switch recombination. J Exp Med 2021; 218:e20201422. [PMID: 34586363 PMCID: PMC8485858 DOI: 10.1084/jem.20201422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 02/12/2021] [Accepted: 09/09/2021] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs (miRNAs, miRs) regulate cell fate decisions by post-transcriptionally tuning networks of mRNA targets. We used miRNA-directed pathway discovery to reveal a regulatory circuit that influences Ig class switch recombination (CSR). We developed a system to deplete mature, activated B cells of miRNAs, and performed a rescue screen that identified the miR-221/222 family as a positive regulator of CSR. Endogenous miR-221/222 regulated B cell CSR to IgE and IgG1 in vitro, and miR-221/222-deficient mice exhibited defective IgE production in allergic airway challenge and polyclonal B cell activation models in vivo. We combined comparative Ago2-HITS-CLIP and gene expression analyses to identify mRNAs bound and regulated by miR-221/222 in primary B cells. Interrogation of these putative direct targets uncovered functionally relevant downstream genes. Genetic depletion or pharmacological inhibition of Foxp1 and Arid1a confirmed their roles as key modulators of CSR to IgE and IgG1.
Collapse
Affiliation(s)
- Eric J. Wigton
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA
| | - Yohei Mikami
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Rockville, MD
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ryan J. McMonigle
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Carlos A. Castellanos
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA
| | - Adam K. Wade-Vallance
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA
| | - Simon K. Zhou
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA
| | - Robin Kageyama
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA
| | - Adam Litterman
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA
| | - Suparna Roy
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Department of Dermatology, University of California, San Francisco, San Francisco, CA
| | - Daisuke Kitamura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Emily C. Dykhuizen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN
| | - Christopher D.C. Allen
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA
| | - Hui Hu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - John J. O’Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Rockville, MD
| | - K. Mark Ansel
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
41
|
The Potential Role of Cytokines and Growth Factors in the Pathogenesis of Alzheimer's Disease. Cells 2021; 10:cells10102790. [PMID: 34685770 PMCID: PMC8534363 DOI: 10.3390/cells10102790] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most prominent neurodegenerative diseases, which impairs cognitive function in afflicted individuals. AD results in gradual decay of neuronal function as a consequence of diverse degenerating events. Several neuroimmune players (such as cytokines and growth factors that are key players in maintaining CNS homeostasis) turn aberrant during crosstalk between the innate and adaptive immunities. This aberrance underlies neuroinflammation and drives neuronal cells toward apoptotic decline. Neuroinflammation involves microglial activation and has been shown to exacerbate AD. This review attempted to elucidate the role of cytokines, growth factors, and associated mechanisms implicated in the course of AD, especially with neuroinflammation. We also evaluated the propensities and specific mechanism(s) of cytokines and growth factors impacting neuron upon apoptotic decline and further shed light on the availability and accessibility of cytokines across the blood-brain barrier and choroid plexus in AD pathophysiology. The pathogenic and the protective roles of macrophage migration and inhibitory factors, neurotrophic factors, hematopoietic-related growth factors, TAU phosphorylation, advanced glycation end products, complement system, and glial cells in AD and neuropsychiatric pathology were also discussed. Taken together, the emerging roles of these factors in AD pathology emphasize the importance of building novel strategies for an effective therapeutic/neuropsychiatric management of AD in clinics.
Collapse
|
42
|
Ortega-Rivera O, Shin MD, Chen A, Beiss V, Moreno-Gonzalez MA, Lopez-Ramirez MA, Reynoso M, Wang H, Hurst BL, Wang J, Pokorski JK, Steinmetz NF. Trivalent Subunit Vaccine Candidates for COVID-19 and Their Delivery Devices. J Am Chem Soc 2021; 143:14748-14765. [PMID: 34490778 PMCID: PMC8442557 DOI: 10.1021/jacs.1c06600] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Indexed: 02/06/2023]
Abstract
The COVID-19 pandemic highlights the need for platform technologies enabling rapid development of vaccines for emerging viral diseases. The current vaccines target the SARS-CoV-2 spike (S) protein and thus far have shown tremendous efficacy. However, the need for cold-chain distribution, a prime-boost administration schedule, and the emergence of variants of concern (VOCs) call for diligence in novel SARS-CoV-2 vaccine approaches. We studied 13 peptide epitopes from SARS-CoV-2 and identified three neutralizing epitopes that are highly conserved among the VOCs. Monovalent and trivalent COVID-19 vaccine candidates were formulated by chemical conjugation of the peptide epitopes to cowpea mosaic virus (CPMV) nanoparticles and virus-like particles (VLPs) derived from bacteriophage Qβ. Efficacy of this approach was validated first using soluble vaccine candidates as solo or trivalent mixtures and subcutaneous prime-boost injection. The high thermal stability of our vaccine candidates allowed for formulation into single-dose injectable slow-release polymer implants, manufactured by melt extrusion, as well as microneedle (MN) patches, obtained through casting into micromolds, for prime-boost self-administration. Immunization of mice yielded high titers of antibodies against the target epitope and S protein, and data confirms that antibodies block receptor binding and neutralize SARS-CoV and SARS-CoV-2 against infection of human cells. We present a nanotechnology vaccine platform that is stable outside the cold-chain and can be formulated into delivery devices enabling single administration or self-administration. CPMV or Qβ VLPs could be stockpiled, and epitopes exchanged to target new mutants or emergent diseases as the need arises.
Collapse
Affiliation(s)
- Oscar
A. Ortega-Rivera
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Matthew D. Shin
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Angela Chen
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Veronique Beiss
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Miguel A. Moreno-Gonzalez
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Miguel A. Lopez-Ramirez
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Maria Reynoso
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322, United States
| | - Hong Wang
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Brett L. Hurst
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322, United States
| | - Joseph Wang
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Jonathan K. Pokorski
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Nicole F. Steinmetz
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| |
Collapse
|
43
|
Song G, Yang H, Shen N, Pham P, Brown B, Lin X, Hong Y, Sinu P, Cai J, Li X, Leon M, Gordon MN, Morgan D, Zhang S, Cao C. An Immunomodulatory Therapeutic Vaccine Targeting Oligomeric Amyloid-β. J Alzheimers Dis 2021; 77:1639-1653. [PMID: 32925044 DOI: 10.3233/jad-200413] [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] [Indexed: 02/06/2023]
Abstract
BACKGROUND Aging is considered the most important risk factor for Alzheimer's disease (AD). Recent research supports the theory that immunotherapy targeting the "oligomeric" forms of amyloid-β (Aβ) may halt the progression of AD. However, previous clinical trial of the vaccine against Aβ, called AN1792, was suspended due to cases of meningoencephalitis in patients. OBJECTIVE To develop a peptide sensitized dendritic cells (DCs) vaccine that would target oligomer Aβ and prevent an autoimmune response. METHODS Double transgenic APPswe/PS1ΔE9 (Tg) and C57BL/6J control mice were used in this study. Cytokine expression profile detection, characterization of antisera, brain GSK-3β, LC3 expression, and spatial working memory testing before and post-vaccination were obtained. RESULTS Epitope prediction indicated that E22W42 could generate 13 new T cell epitopes which can strengthen immunity in aged subjects and silence several T cell epitopes of the wild type Aβ. The silenced T cell epitope could help avoid the autoimmune response that was seen in some patients of the AN-1792 vaccine. The E22W42 not only helped sensitize bone marrow-derived DCs for the development of an oligomeric Aβ-specific antibody, but also delayed memory impairment in the APP/PS1 mouse model. Most importantly, this E22W42 peptide will not alter the DC's natural immunomodulatory properties. CONCLUSION The E22W42 vaccine is possibly safer for patients with impaired immune systems. Since there is increasing evidence that oligomeric form of Aβ are the toxic species to neurons, the E22W42 antibody's specificity for these "oligomeric" Aβ species could provide the opportunity to produce some clinical benefits in AD subjects.
Collapse
Affiliation(s)
- Ge Song
- Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Haiqiang Yang
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Ning Shen
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Phillip Pham
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Breanna Brown
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xiaoyang Lin
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Yuzhu Hong
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Paul Sinu
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Jianfeng Cai
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Xiaopeng Li
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Michael Leon
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Marcia N Gordon
- Department of Translational Neuroscience, College of Medicine, Michigan State University, Grand Rapids, MI, USA
| | - David Morgan
- Department of Translational Neuroscience, College of Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Sai Zhang
- Institution of Brain Trauma and Neurology Disease, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Chuanhai Cao
- College of Arts and Science, University of South Florida, Tampa, FL, USA.,Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA.,College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
44
|
Inhibition of elastase enhances the adjuvanticity of alum and promotes anti-SARS-CoV-2 systemic and mucosal immunity. Proc Natl Acad Sci U S A 2021; 118:2102435118. [PMID: 34353890 PMCID: PMC8403952 DOI: 10.1073/pnas.2102435118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We report that suppression of the serine protease elastase reshapes innate responses induced by injected vaccines containing alum adjuvant. This reprogramming improves the induction of protective antibodies in the bloodstream and stimulates innate signals, which support the development of antibody responses in mucosal tissues. Our findings identify elastase as the innate regulator that blunts the adjuvant activity of alum. They also demonstrate that vaccination via mucosal routes is not an absolute requirement for antibody responses in mucosal tissues and secretions. Supplementation of an alum-based vaccine containing SARS-CoV-2 spike protein subunit 1 as antigen increased anti–SARS-CoV-2 immunity in the blood and mucosal secretions in mice. Thus, this strategy could help in the development of future protein-based vaccines against SARS-CoV-2. Alum, used as an adjuvant in injected vaccines, promotes T helper 2 (Th2) and serum antibody (Ab) responses. However, it fails to induce secretory immunoglobulin (Ig) A (SIgA) in mucosal tissues and is poor in inducing Th1 and cell-mediated immunity. Alum stimulates interleukin 1 (IL-1) and the recruitment of myeloid cells, including neutrophils. We investigated whether neutrophil elastase regulates the adjuvanticity of alum, and whether a strategy targeting neutrophil elastase could improve responses to injected vaccines. Mice coadministered a pharmacological inhibitor of elastase, or lacking elastase, developed high-affinity serum IgG and IgA antibodies after immunization with alum-adsorbed protein vaccines, including the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2). These mice also developed broader antigen-specific CD4+ T cell responses, including high Th1 and T follicular helper (Tfh) responses. Interestingly, in the absence of elastase activity, mucosal SIgA responses were induced after systemic immunization with alum as adjuvant. Importantly, lack or suppression of elastase activity enhanced the magnitude of anti–SARS-CoV-2 spike subunit 1 (S1) antibodies, and these antibodies reacted with the same epitopes of spike 1 protein as sera from COVID-19 patients. Therefore, suppression of neutrophil elastase could represent an attractive strategy for improving the efficacy of alum-based injected vaccines for the induction of broad immunity, including mucosal immunity.
Collapse
|
45
|
Ortega-Rivera OA, Pokorski JK, Steinmetz NF. A single-dose, implant-based, trivalent virus-like particle vaccine against "cholesterol checkpoint" proteins. ADVANCED THERAPEUTICS 2021; 4:2100014. [PMID: 34541299 PMCID: PMC8447230 DOI: 10.1002/adtp.202100014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Indexed: 01/23/2023]
Abstract
Cardiovascular disease is the number one cause of death globally. Lowering cholesterol levels in plasma is the mainstay therapy; however lifelong treatment and adverse effects call for improved therapeutic interventions. We developed a trivalent vaccine candidate targeting proprotein convertase subtilisin/kexin-9 (PCSK9), apolipoprotein B (ApoB), and cholesteryl ester transfer protein (CETP). Vaccine candidates were developed using bacteriophage Qβ-based virus-like particles (VLPs) displaying antigens of PCKS9, ApoB, and CETP, respectively. Vaccine candidate mixtures were formulated as slow-release PLGA:VLP implants using hot-melt extrusion. The delivery of the trivalent vaccine candidate via the implant produced antibodies against the cholesterol checkpoint proteins at levels comparable to a three-dose injection schedule with soluble mixtures. The reduction in PCSK9 and ApoB levels in plasma, inhibition of CETP (in vitro), and total plasma cholesterol decrease was achieved. All-together, we present a platform technology for a single-dose multi-target vaccination platform targeting cholesterol checkpoint proteins.
Collapse
Affiliation(s)
- Oscar A. Ortega-Rivera
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
| | - Jonathan K. Pokorski
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Institute for Materials Discovery and Design, University of California-San Diego, La Jolla CA 92039, USA
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla CA 92039, USA
- Institute for Materials Discovery and Design, University of California-San Diego, La Jolla CA 92039, USA
- Department of Bioengineering, University of California-San Diego, La Jolla CA 92039, USA
- Department of Radiology, University of California-San Diego, La Jolla CA 92039, USA
- Moores Cancer Center, University of California-San Diego, La Jolla CA 92039, USA
| |
Collapse
|
46
|
Zhai C, Zheng XJ, Song C, Ye XS. Synthesis and immunological evaluation of N-acyl modified Globo H derivatives as anticancer vaccine candidates. RSC Med Chem 2021; 12:1239-1243. [PMID: 34355188 PMCID: PMC8292959 DOI: 10.1039/d1md00067e] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Globo H is a tumor-associated carbohydrate antigen (TACA), which serves as a valuable target for antitumor vaccine or cancer immunotherapies. However, most TACAs are T-cell-independent, and they cannot induce powerful immune response due to their poor immunogenicity. To address this problem, herein, several Globo H analogues with modification on the N-acyl group were prepared through a preactivation-based glycosylation strategy from the non-reducing end to the reducing end. These modified Globo H derivatives were then conjugated with carrier protein CRM197 to form glycoconjugates as anticancer vaccine candidates, which were used in combination with adjuvant glycolipid C34 for immunological studies. The immunological effects of these synthetic vaccine candidates were evaluated on Balb/c mice. The results showed that the fluorine-modified N-acyl Globo H conjugates can induce higher titers of IgG antibodies that can recognize the naturally occurring Globo H antigen on the surface of cancer cells and can eliminate cancer cells in the presence of a complement, indicating the potential of these synthetic glycoconjugates as anticancer vaccine candidates.
Collapse
Affiliation(s)
- Canjia Zhai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Xue Yuan Rd No. 38 Beijing 100191 China
| | - Xiu-Jing Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Xue Yuan Rd No. 38 Beijing 100191 China
| | - Chengcheng Song
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Xue Yuan Rd No. 38 Beijing 100191 China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Xue Yuan Rd No. 38 Beijing 100191 China
| |
Collapse
|
47
|
Wade-Vallance AK, Allen CDC. Intrinsic and extrinsic regulation of IgE B cell responses. Curr Opin Immunol 2021; 72:221-229. [PMID: 34216934 DOI: 10.1016/j.coi.2021.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/26/2021] [Accepted: 06/03/2021] [Indexed: 01/23/2023]
Abstract
Stringent regulation of IgE antibody production is critical for constraining allergic responses. This review discusses recent advances in understanding cell-intrinsic and extrinsic mechanisms that regulate the genesis and fate of IgE B cells. B cell-intrinsic regulation of IgE is orchestrated by the IgE B Cell Receptor (BCR). Through its antigen-independent signaling and low surface expression, the IgE BCR drives IgE B cells to differentiate into short-lived plasma cells and/or undergo apoptosis, restricting IgE-expressing cells from entering long-lived compartments. The pivotal extrinsic regulators of IgE responses are T follicular helper cells (TFH). TFH produce IL-4 and IL-21, which, respectively, are the major activating and inhibitory cytokines for IgE class-switching. Other newly identified T follicular subsets also contribute to IgE regulation. Although IgE responses are normally constrained, recent studies suggest that specific conditions can induce the formation of IgE responses with enhanced affinity or longevity, effectively 'breaking the rules' of IgE regulation.
Collapse
Affiliation(s)
- Adam K Wade-Vallance
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
48
|
Khatun MM, Islam MA, Baek BK. In Vitro and In Vivo IFN-γ and IL-10 Measurement in Experimental Brucella abortus Biotype 1 Infection in Sprague-Dawley Rats. Vector Borne Zoonotic Dis 2021; 21:579-585. [PMID: 34077683 DOI: 10.1089/vbz.2020.2738] [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] [Indexed: 11/12/2022] Open
Abstract
The immune response to Brucella abortus mainly depends on antigen-specific T cell activation, CD4+ and CD8+ T cells, and Brucella-specific humoral response. Protective immune response against Brucella infection has not been performed in the Sprague-Dawley (SD) rat model. We measured bacterial kinetics in addition to in vivo and in vitro interferon gamma (IFN-γ) and interleukin-10 (IL-10) production against crude Brucella protein in the SD rats at different days of postinfection with B. abortus biotype 1 by indirect enzyme-linked immunosorbent assay. Forty SD rats were inoculated intraperitoneally with 0.1 mL sterile injectable pyrogen-free solution containing 1 × 1010 colony-forming units/mL of B. abortus biotype 1 obtained from cattle in Korea. Four rats were used as uninfected control. Serum IFN-γ level at 3 and 7 days postinfection were significantly higher (p > 0.001) compared with the IL-10 level. On the contrary, serum IL-10 levels were observed significantly higher at 21 and 28 days postinfection compared with the serum IFN-γ levels (p < 0.001). The production of IFN-γ by spleen cells was significantly higher at 7 and 14 days postinfection compared with IL-10 (p < 0.001). On the contrary, IL-10 productions were found to be significantly higher at 21, 28, 35, and 42 days postinfection compared with IFN-γ (p < 0.001). The presence of B. abortus in blood was marked till 5 weeks of infection, throughout the experiment in case of spleen, and no bacteria were isolated from the kidney and liver at 6 weeks postinfection. The in vivo and in vitro IFN-γ and IL-10 measurement in our study reported that B. abortus infection in rats primarily educe T helper (Th)1-dominant immune response in acute infection accompanied by Th2-dominant immune response in chronic infection.
Collapse
Affiliation(s)
- Mst Minara Khatun
- Department of Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Jeonju, Republic of Korea.,Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Ariful Islam
- Department of Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Jeonju, Republic of Korea.,Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Byeong Kirl Baek
- Department of Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
49
|
Guzelj S, Nabergoj S, Gobec M, Pajk S, Klančič V, Slütter B, Frkanec R, Štimac A, Šket P, Plavec J, Mlinarič-Raščan I, Jakopin Ž. Structural Fine-Tuning of Desmuramylpeptide NOD2 Agonists Defines Their In Vivo Adjuvant Activity. J Med Chem 2021; 64:7809-7838. [PMID: 34043358 PMCID: PMC8279416 DOI: 10.1021/acs.jmedchem.1c00644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
We
report on the design, synthesis, and biological evaluation of
a series of nucleotide-binding oligomerization-domain-containing protein
2 (NOD2) desmuramylpeptide agonists with improved in vitro and in vivo adjuvant properties. We identified
two promising compounds: 68, a potent nanomolar in vitro NOD2 agonist, and the more lipophilic 75, which shows superior adjuvant activity in vivo. Both compounds had immunostimulatory effects on peripheral blood
mononuclear cells at the protein and transcriptional levels, and augmented
dendritic-cell-mediated activation of T cells, while 75 additionally enhanced the cytotoxic activity of peripheral blood
mononuclear cells against malignant cells. The C18 lipophilic
tail of 75 is identified as a pivotal structural element
that confers in vivo adjuvant activity in conjunction
with a liposomal delivery system. Accordingly, liposome-encapsulated 75 showed promising adjuvant activity in mice, surpassing
that of muramyl dipeptide, while achieving a more balanced Th1/Th2
immune response, thus highlighting its potential as a vaccine adjuvant.
Collapse
Affiliation(s)
- Samo Guzelj
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Sanja Nabergoj
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Martina Gobec
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Stane Pajk
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Veronika Klančič
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Bram Slütter
- Div. BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Ruža Frkanec
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Adela Štimac
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Primož Šket
- Slovenian NMR Centre, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia
| | - Janez Plavec
- Slovenian NMR Centre, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia
| | | | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
50
|
Tada R, Suzuki H, Ogasawara M, Yamanaka D, Adachi Y, Kunisawa J, Negishi Y. Polymeric Caffeic Acid Acts as a Nasal Vaccine Formulation against Streptococcus pneumoniae Infections in Mice. Pharmaceutics 2021; 13:pharmaceutics13040585. [PMID: 33923897 PMCID: PMC8073337 DOI: 10.3390/pharmaceutics13040585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/17/2021] [Accepted: 04/17/2021] [Indexed: 11/16/2022] Open
Abstract
Infectious diseases are the second leading cause of death worldwide, highlighting the importance of the development of a novel and improved strategy for fighting pathogenic microbes. Streptococcus pneumoniae is a highly pathogenic bacteria that causes pneumonia with high mortality rates, especially in children and elderly individuals. To solve these issues, a mucosal vaccine system would be the best solution for the prevention and treatment of these diseases. We have recently reported that enzymatically polymerized caffeic acid (pCA) acts as a mucosal adjuvant when co-administered with antigenic proteins via the nasal route. Moreover, the sources of caffeic acid and horseradish peroxidase are ingredients found commonly in coffee beans and horseradish, respectively. In this study, we aimed to develop a pneumococcal nasal vaccine comprising pneumococcal surface protein A (PspA) and pCA as the mucosal adjuvant. Intranasal immunization with PspA and pCA induced the production of PspA-specific antibody responses in the mucosal and systemic compartments. Furthermore, the protective effects were tested in a murine model of S. pneumoniae infection. Intranasal vaccination conferred antigen-dependent protective immunity against a lethal infection of S. pneumoniae. In conclusion, pCA is useful as a serotype-independent universal nasal pneumococcal vaccine formulation.
Collapse
Affiliation(s)
- Rui Tada
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (M.O.); (Y.N.)
- Correspondence: ; Tel.: +81-42-676-3219
| | - Hidehiko Suzuki
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; (H.S.); (J.K.)
| | - Miki Ogasawara
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (M.O.); (Y.N.)
| | - Daisuke Yamanaka
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (D.Y.); (Y.A.)
| | - Yoshiyuki Adachi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (D.Y.); (Y.A.)
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan; (H.S.); (J.K.)
- Division of Mucosal Immunology and International Research and Development Center for Mucosal Vaccines, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (M.O.); (Y.N.)
| |
Collapse
|