1
|
Su Z, Boucetta H, Shao J, Huang J, Wang R, Shen A, He W, Xu ZP, Zhang L. Next-generation aluminum adjuvants: Immunomodulatory layered double hydroxide NanoAlum reengineered from first-line drugs. Acta Pharm Sin B 2024; 14:4665-4682. [PMID: 39664431 PMCID: PMC11628803 DOI: 10.1016/j.apsb.2024.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 12/13/2024] Open
Abstract
Aluminum adjuvants (Alum), approved by the US Food and Drug Administration, have been extensively used in vaccines containing recombinant antigens, subunits of pathogens, or toxins for almost a century. While Alums typically elicit strong humoral immune responses, their ability to induce cellular and mucosal immunity is limited. As an alternative, layered double hydroxide (LDH), a widely used antacid, has emerged as a novel class of potent nano-aluminum adjuvants (NanoAlum), demonstrating advantageous physicochemical properties, biocompatibility and adjuvanticity in both humoral and cellular immune responses. In this review, we summarize and compare the advantages and disadvantages of Alum and NanoAlum in these properties and their performance as adjuvants. Moreover, we propose the key features for ideal adjuvants and demonstrate that LDH NanoAlum is a promising candidate by summarizing its current progress in immunotherapeutic cancer treatments. Finally, we conclude the review by offering our integrated perspectives about the remaining challenges and future directions for NanoAlum's application in preclinical/clinical settings.
Collapse
Affiliation(s)
- Zhenwei Su
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Hamza Boucetta
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jiahui Shao
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Jinling Huang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ran Wang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Aining Shen
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhi Ping Xu
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Lingxiao Zhang
- Interdisciplinary Nanoscience Center (INANO), Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
2
|
Gogoi H, Mani R, Bhatnagar R. Re-inventing traditional aluminum-based adjuvants: Insight into a century of advancements. Int Rev Immunol 2024; 44:58-81. [PMID: 39310923 DOI: 10.1080/08830185.2024.2404095] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 06/01/2023] [Accepted: 08/31/2024] [Indexed: 02/22/2025]
Abstract
Aluminum salt-based adjuvants like alum, alhydrogel and Adju-Phos are by far the most favored clinically approved vaccine adjuvants. They have demonstrated excellent safety profile and currently used in vaccines against diphtheria, tetanus, pertussis, hepatitis B, anthrax etc. These vaccinations cause minimal side effects like local inflammation at the injection site. Aluminum salt-based adjuvants primarily stimulate CD4+ T cells and B cell mediated Th2 immune response leading to generate a robust antibody response. In this review article, we have compiled the role of physio-chemical role of the two commonly used aluminum salt-based adjuvants alhydrogel and Adju-Phos, and the effect of surface properties, buffer composition, and adjuvant dosage on the immune response. After being studied for almost a century, researchers have come up with various mechanism by which these aluminum adjuvants activate the immune system. Firstly, we have covered the initial works of Glenny and his "repository effect" which paved the work for his successors to explore the involvement of cytokines, chemokines, recruitment of innate immune cells, enhanced antigen uptake by antigen presenting cells, and formation of NLRP3 inflammasome complex in mediating the immune response. It has been reported that aluminum adjuvants activate multiple immunological pathways which synergistically activates the immune system. We later discuss the recent developments in nanotechnology-based preparations of next generation aluminum based adjuvants which has enabled precise size control and morphology of the traditional aluminum adjuvants thereby manipulating the immune response as per our desire.
Collapse
Affiliation(s)
- Himanshu Gogoi
- Amity Institute of Microbial Technology, Amity University Rajasthan, Jaipur, India
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, Faridabad, Haryana, India
| | - Rajesh Mani
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Department of Microbiology, Immunology and Molecular Genetics, University Kentucky College of Medicine, Lexington, KY, USA
| | - Rakesh Bhatnagar
- Amity Institute of Microbial Technology, Amity University Rajasthan, Jaipur, India
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
3
|
Gao X, Wang X, Li S, Saif Ur Rahman M, Xu S, Liu Y. Nanovaccines for Advancing Long-Lasting Immunity against Infectious Diseases. ACS NANO 2023; 17:24514-24538. [PMID: 38055649 DOI: 10.1021/acsnano.3c07741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Infectious diseases, particularly life-threatening pathogens such as small pox and influenza, have substantial implications on public health and global economies. Vaccination is a key approach to combat existing and emerging pathogens. Immunological memory is an essential characteristic used to evaluate vaccine efficacy and durability and the basis for the long-term effects of vaccines in protecting against future infections; however, optimizing the potency, improving the quality, and enhancing the durability of immune responses remains challenging and a focus for research involving investigation of nanovaccine technologies. In this review, we describe how nanovaccines can address the challenges for conventional vaccines in stimulating adaptive immune memory responses to protect against reinfection. We discuss protein and nonprotein nanoparticles as useful antigen platforms, including those with highly ordered and repetitive antigen array presentation to enhance immunogenicity through cross-linking with multiple B cell receptors, and with a focus on antigen properties. In addition, we describe how nanoadjuvants can improve immune responses by providing enhanced access to lymph nodes, lymphnode targeting, germinal center retention, and long-lasting immune response generation. Nanotechnology has the advantage to facilitate vaccine induction of long-lasting immunity against infectious diseases, now and in the future.
Collapse
Affiliation(s)
- Xinglong Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | | | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P.R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
4
|
Zhang Y, Hardy LC, Kapita CM, Hall JA, Arbeeva L, Campbell E, Urban JF, Belkaid Y, Nagler CR, Iweala OI. Intestinal Helminth Infection Impairs Oral and Parenteral Vaccine Efficacy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:389-402. [PMID: 37272847 PMCID: PMC10524302 DOI: 10.4049/jimmunol.2300084] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023]
Abstract
The impact of endemic parasitic infection on vaccine efficacy is an important consideration for vaccine development and deployment. We have examined whether intestinal infection with the natural murine helminth Heligmosomoides polygyrus bakeri alters Ag-specific Ab and cellular immune responses to oral and parenteral vaccination in mice. Oral vaccination of mice with a clinically relevant, live, attenuated, recombinant Salmonella vaccine expressing chicken egg OVA (Salmonella-OVA) induced the accumulation of activated, OVA-specific T effector cells rather than OVA-specific regulatory T cells in the GALT. Intestinal helminth infection significantly reduced Th1-skewed Ab responses to oral vaccination with Salmonella-OVA. Activated, adoptively transferred, OVA-specific CD4+ T cells accumulated in draining mesenteric lymph nodes of vaccinated mice, regardless of their helminth infection status. However, helminth infection increased the frequencies of adoptively transferred OVA-specific CD4+ T cells producing IL-4 and IL-10 in the mesenteric lymph node. Ab responses to the oral Salmonella-OVA vaccine were reduced in helminth-free mice adoptively transferred with OVA-specific CD4+ T cells harvested from mice with intestinal helminth infection. Intestinal helminth infection also significantly reduced Th2-skewed Ab responses to parenteral vaccination with OVA adsorbed to alum. These findings suggest that vaccine-specific CD4+ T cells induced in the context of helminth infection retain durable immunomodulatory properties and may promote blunted Ab responses to vaccination. They also underscore the potential need to treat parasitic infection before mass vaccination campaigns in helminth-endemic areas.
Collapse
Affiliation(s)
- Yugen Zhang
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599
| | - LaKeya C. Hardy
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599
| | - Camille M. Kapita
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
| | - Jason A. Hall
- National Institute of Allergy and Infectious Diseases Microbiome Program and Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, Center for Human Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Liubov Arbeeva
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
| | - Evelyn Campbell
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637
| | - Joseph F. Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory and Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, 10300 Baltimore Avenue BLDG 307-C BARC-East, Beltsville, MD, 20705
| | - Yasmine Belkaid
- National Institute of Allergy and Infectious Diseases Microbiome Program and Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, Center for Human Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Cathryn R. Nagler
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637
- Center for Immunology and Inflammatory Disease, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Charlestown, MA 02129
| | - Onyinye I. Iweala
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599
- Center for Immunology and Inflammatory Disease, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Charlestown, MA 02129
| |
Collapse
|
5
|
Rivera CE, Zhou Y, Chupp DP, Yan H, Fisher AD, Simon R, Zan H, Xu Z, Casali P. Intrinsic B cell TLR-BCR linked coengagement induces class-switched, hypermutated, neutralizing antibody responses in absence of T cells. SCIENCE ADVANCES 2023; 9:eade8928. [PMID: 37115935 PMCID: PMC10146914 DOI: 10.1126/sciadv.ade8928] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/23/2023] [Indexed: 05/03/2023]
Abstract
Maturation of antibody responses entails somatic hypermutation (SHM), class-switch DNA recombination (CSR), plasma cell differentiation, and generation of memory B cells, and it is thought to require T cell help. We showed that B cell Toll-like receptor 4 (TLR4)-B cell receptor (BCR) (receptor for antigen) coengagement by 4-hydroxy-3-nitrophenyl acetyl (NP)-lipopolysaccharide (LPS) (Escherichia coli lipid A polysaccharide O-antigen) or TLR5-BCR coengagement by Salmonella flagellin induces mature antibody responses to NP and flagellin in Tcrβ-/-Tcrδ-/- and NSG/B mice. TLR-BCR coengagement required linkage of TLR and BCR ligands, "linked coengagement." This induced B cell CSR/SHM, germinal center-like differentiation, clonal expansion, intraconal diversification, plasma cell differentiation, and an anamnestic antibody response. In Tcrβ-/-Tcrδ-/- mice, linked coengagement of TLR4-BCR by LPS or TLR5-BCR by flagellin induced protective antibodies against E. coli or Salmonella Typhimurium. Our findings unveiled a critical role of B cell TLRs in inducing neutralizing antibody responses, including those to microbial pathogens, without T cell help.
Collapse
Affiliation(s)
- Carlos E. Rivera
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Yulai Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Daniel P. Chupp
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Hui Yan
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Amanda D. Fisher
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Raphael Simon
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hong Zan
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Zhenming Xu
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Paolo Casali
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
- Department of Medicine, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
6
|
Bissa M, Kim S, Galli V, Fourati S, Sarkis S, Arakelyan A, de Castro IS, Rahman MA, Fujiwara S, Vaccari M, Tomalka JA, Stamos JD, Schifanella L, Gorini G, Moles R, Gutowska A, Ferrari G, Lobanov A, Montefiori DC, Nelson GW, Cam MC, Chakhtoura M, Haddad EK, Doster MN, McKinnon K, Brown S, Venzon DJ, Choo-Wosoba H, Breed MW, Killoran KE, Kramer J, Margolis L, Sekaly RP, Hager GL, Franchini G. HIV vaccine candidate efficacy in female macaques mediated by cAMP-dependent efferocytosis and V2-specific ADCC. Nat Commun 2023; 14:575. [PMID: 36732510 PMCID: PMC9894672 DOI: 10.1038/s41467-023-36109-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/15/2023] [Indexed: 02/04/2023] Open
Abstract
The development of an effective vaccine to protect against HIV acquisition will be greatly bolstered by in-depth understanding of the innate and adaptive responses to vaccination. We report here that the efficacy of DNA/ALVAC/gp120/alum vaccines, based on V2-specific antibodies mediating apoptosis of infected cells (V2-ADCC), is complemented by efferocytosis, a cyclic AMP (cAMP)-dependent antiphlogistic engulfment of apoptotic cells by CD14+ monocytes. Central to vaccine efficacy is the engagement of the CCL2/CCR2 axis and tolerogenic dendritic cells producing IL-10 (DC-10). Epigenetic reprogramming in CD14+ cells of the cyclic AMP/CREB pathway and increased systemic levels of miRNA-139-5p, a negative regulator of expression of the cAMP-specific phosphodiesterase PDE4D, correlated with vaccine efficacy. These data posit that efferocytosis, through the prompt and effective removal of apoptotic infected cells, contributes to vaccine efficacy by decreasing inflammation and maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Massimiliano Bissa
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA.
| | - Sohyoung Kim
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Veronica Galli
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Slim Fourati
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Emory University, Atlanta, GA, USA
| | - Sarkis Sarkis
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Anush Arakelyan
- Section on Intercellular Interactions, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | | | - Mohammad Arif Rahman
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Saori Fujiwara
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Monica Vaccari
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Jeffrey A Tomalka
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Emory University, Atlanta, GA, USA
| | - James D Stamos
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Luca Schifanella
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Giacomo Gorini
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Ramona Moles
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Anna Gutowska
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Guido Ferrari
- Division of Surgical Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Alexei Lobanov
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David C Montefiori
- Division of Surgical Sciences, Duke University School of Medicine, Durham, NC, USA
| | - George W Nelson
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Margaret C Cam
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Marita Chakhtoura
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Elias K Haddad
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Melvin N Doster
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
| | - Katherine McKinnon
- Vaccine Branch Flow Cytometry Core, National Cancer Institute, Bethesda, MD, USA
| | - Sophia Brown
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA
- Vaccine Branch Flow Cytometry Core, National Cancer Institute, Bethesda, MD, USA
| | - David J Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Matthew W Breed
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD, USA
| | - Kristin E Killoran
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD, USA
| | - Joshua Kramer
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD, USA
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Rafick P Sekaly
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Emory University, Atlanta, GA, USA
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
7
|
Prince L, Martín-Faivre L, Villeret B, Sanchez-Guzman D, Le Guen P, Sallenave JM, Garcia-Verdugo I. Eosinophils Recruited during Pulmonary Vaccination Regulate Mucosal Antibody Production. Am J Respir Cell Mol Biol 2023; 68:186-200. [PMID: 36194580 DOI: 10.1165/rcmb.2022-0236oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Eosinophils have been previously shown to be able to regulate early humoral responses during systemic vaccination. Here we investigated the role of eosinophils during pulmonary vaccination, comparing vaccine-induced responses in eosinophil-deficient (ΔdblGATA) and wild-type mice using a Th2 adjuvant. We observed that eosinophils were needed to induce a complete vaccine response, thereby eliciting specific antibody-secreting plasma cells in the regional lymph nodes and antibody secretion in the BAL at the early stage of the immune response. Reintroduction of eosinophils in the lungs of ΔdblGATA mice during the priming stage enhanced both specific IgM and IgG plasma cells but not specific IgA plasma cells. Upon vaccination, eosinophils migrated to the lungs and secreted cytokines involved in B-cell activation, which might promote antibody production. Importantly, however, the absence of eosinophils did not impair late immune responses in a prime/boost protocol because, in that setup, we uncovered a compensating mechanism involving a Th17 pathway. In conclusion, our data demonstrate for the first time a new role for eosinophils during lung mucosal vaccination, whereby they accelerate early immune responses (IgM and IgG) while regulating IgA production at the late stages.
Collapse
Affiliation(s)
- Lisa Prince
- INSERM U1152, Physiopathologie et épidémiologie des maladies respiratoires, Université Paris Cité, Paris, France
| | - Lydie Martín-Faivre
- INSERM U1152, Physiopathologie et épidémiologie des maladies respiratoires, Université Paris Cité, Paris, France
| | - Bérengère Villeret
- INSERM U1152, Physiopathologie et épidémiologie des maladies respiratoires, Université Paris Cité, Paris, France
| | - Daniel Sanchez-Guzman
- INSERM U1152, Physiopathologie et épidémiologie des maladies respiratoires, Université Paris Cité, Paris, France
| | - Pierre Le Guen
- INSERM U1152, Physiopathologie et épidémiologie des maladies respiratoires, Université Paris Cité, Paris, France
| | - Jean-Michel Sallenave
- INSERM U1152, Physiopathologie et épidémiologie des maladies respiratoires, Université Paris Cité, Paris, France
| | - Ignacio Garcia-Verdugo
- INSERM U1152, Physiopathologie et épidémiologie des maladies respiratoires, Université Paris Cité, Paris, France
| |
Collapse
|
8
|
August A, Marichal T. Eosinophils and Lung Mucosal Antibody Production: Is Location the Key? Am J Respir Cell Mol Biol 2023; 68:124-126. [PMID: 36306503 PMCID: PMC9986563 DOI: 10.1165/rcmb.2022-0410ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Avery August
- Department of Microbiology & Immunology Cornell University Ithaca, New York
| | | |
Collapse
|
9
|
A Nonadjuvanted Whole-Inactivated Pneumococcal Vaccine Induces Multiserotype Opsonophagocytic Responses Mediated by Noncapsule-Specific Antibodies. mBio 2022; 13:e0236722. [PMID: 36125268 PMCID: PMC9600166 DOI: 10.1128/mbio.02367-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Streptococcus pneumoniae (Spn) remains a major cause of global mortality, with extensive antigenic diversity between capsular serotypes that poses an ongoing challenge for vaccine development. Widespread use of pneumococcal conjugate vaccines (PCVs) targeting Spn capsules has greatly reduced infections by vaccine-included serotypes but has led to increased infections by nonincluded serotypes. To date, high cost of PCVs has also limited their usefulness in low-income regions where disease burdens are highest. To overcome these limitations, serotype-independent vaccines are being actively researched. We have developed a whole-cell gamma-irradiated Spn vaccine (termed Gamma-PN) providing serotype-independent protection. We demonstrate that Gamma-PN immunization of mice or rabbits via the clinically relevant intramuscular route induces protein-specific antibodies able to bind numerous nonvaccine encapsulated serotypes, which mediate opsonophagocytic killing and protection against lethal challenges. Gamma-PN induced comparable or superior opsonophagocytic killing assay (OPKA) responses in rabbits to the licensed Prevnar 13 vaccine (PCV13) for vaccine-included serotypes, and a superior response to nonincluded serotypes, including emergent 22F and 35B. Additionally, despite a lower observed reactogenicity, administration of Gamma-PN without adjuvant resulted in higher OPKA responses and improved protection compared to adjuvanted Gamma-PN. To our knowledge, this has not been demonstrated previously for a whole-inactivated Spn vaccine. Eliminating the requirement for adjuvant comes with numerous benefits for clinical applications of this vaccine and poses interesting questions for the inclusion of adjuvant in similar vaccines in development.
Collapse
|
10
|
Howard FHN, Kwan A, Winder N, Mughal A, Collado-Rojas C, Muthana M. Understanding Immune Responses to Viruses-Do Underlying Th1/Th2 Cell Biases Predict Outcome? Viruses 2022; 14:1493. [PMID: 35891472 PMCID: PMC9324514 DOI: 10.3390/v14071493] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 12/15/2022] Open
Abstract
Emerging and re-emerging viral diseases have increased in number and geographical extent during the last decades. Examples include the current COVID-19 pandemic and the recent epidemics of the Chikungunya, Ebola, and Zika viruses. Immune responses to viruses have been well-characterised within the innate and adaptive immunity pathways with the outcome following viral infection predominantly attributed to properties of the virus and circumstances of the infection. Perhaps the belief that the immune system is often considered as a reactive component of host defence, springing into action when a threat is detected, has contributed to a poorer understanding of the inherent differences in an individual's immune system in the absence of any pathology. In this review, we focus on how these host factors (age, ethnicity, underlying pathologies) may skew the T helper cell response, thereby influencing the outcome following viral infection but also whether we can use these inherent biases to predict patients at risk of a deviant response and apply strategies to avoid or overcome them.
Collapse
Affiliation(s)
- Faith H. N. Howard
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK; (A.K.); (N.W.); (A.M.); (C.C.-R.); (M.M.)
| | | | | | | | | | | |
Collapse
|
11
|
Sibille A, Corhay JL, Louis R, Ninane V, Jerusalem G, Duysinx B. Eosinophils and Lung Cancer: From Bench to Bedside. Int J Mol Sci 2022; 23:ijms23095066. [PMID: 35563461 PMCID: PMC9101877 DOI: 10.3390/ijms23095066] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 02/05/2023] Open
Abstract
Eosinophils are rare, multifunctional granulocytes. Their growth, survival, and tissue migration mainly depend on interleukin (IL)-5 in physiological conditions and on IL-5 and IL-33 in inflammatory conditions. Preclinical evidence supports an immunological role for eosinophils as innate immune cells and as agents of the adaptive immune response. In addition to these data, several reports show a link between the outcomes of patients treated with immune checkpoint inhibitors (ICI) for advanced cancers and blood eosinophilia. In this review, we present, in the context of non-small cell lung cancer (NSCLC), the biological properties of eosinophils and their roles in homeostatic and pathological conditions, with a focus on their pro- and anti-tumorigenic effects. We examine the possible explanations for blood eosinophilia during NSCLC treatment with ICI. In particular, we discuss the value of eosinophils as a potential prognostic and predictive biomarker, highlighting the need for stronger clinical data. Finally, we conclude with perspectives on clinical and translational research topics on this subject.
Collapse
Affiliation(s)
- Anne Sibille
- Department of Pulmonology, University Hospital of Liège, Domaine de l’Université B35, 4000 Liège, Belgium; (J.-L.C.); (R.L.); (B.D.)
- Correspondence: ; Tel.: +32-4-3667881
| | - Jean-Louis Corhay
- Department of Pulmonology, University Hospital of Liège, Domaine de l’Université B35, 4000 Liège, Belgium; (J.-L.C.); (R.L.); (B.D.)
| | - Renaud Louis
- Department of Pulmonology, University Hospital of Liège, Domaine de l’Université B35, 4000 Liège, Belgium; (J.-L.C.); (R.L.); (B.D.)
| | - Vincent Ninane
- Department of Pulmonary Medicine, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital of Liège, Domaine de l’Université B35, 4000 Liège, Belgium;
| | - Bernard Duysinx
- Department of Pulmonology, University Hospital of Liège, Domaine de l’Université B35, 4000 Liège, Belgium; (J.-L.C.); (R.L.); (B.D.)
| |
Collapse
|
12
|
Li D, Xu M, Li G, Zheng Y, Zhang Y, Xia D, Wang S, Chen Y. Mg/Al-LDH as a nano-adjuvant for pertussis vaccine: a evaluation compared with aluminum hydroxide adjuvant. NANOTECHNOLOGY 2022; 33:235102. [PMID: 35189608 DOI: 10.1088/1361-6528/ac56f3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
Background. Layered double hydroxide (LDH) has been demonstrated as a highly efficient antigen platform to induce effective and durable immune response. However, whether LDH nanoparticles could act as an adjuvant for pertussis vaccines is still unknown. Here we evaluated the potential of Mg/Al-LDH as a nano-adjuvant to improve immune response against pertussis and compared it with commercial aluminum hydroxide (AH) adjuvant.Method. The Mg/Al-LDH nanoparticles were synthesized by a hydrothermal reaction. The morphology, structure and size of Mg/Al-LDH were characterized by transmission electron microscope, x-ray diffraction and MALVERN particle analysis. The ovalbumin and Pertussis toxin (PTd) was adsorbed to Mg/Al-LDH. The immune response of antigen-LDH complex was evaluated in mice, compared with commercial adjuvant alum. Hematoxylin-eosin staining was used to evaluate the inflammatory response at injection site.Results. The synthetic Mg/Al-LDH nanoparticles showed a typical hexagonal lamellar structure. The average size of synthetic nanoparticles was 102.9 nm with PDI of 0.13 and zeta potential was 44.4 mV. Mg/Al-LDH nanoparticles effectively adsorbed protein antigen and mediated antigen uptake by DC cells. Animal experiments showed that Mg/Al-LDH gave enhancement in anti-pertussis toxin (PTd) humoral immune response, which was considerable to commercial AH adjuvant. Finally, Mg/Al-LDH produced a slighter inflammatory response than AH at injection site and this injury was quickly recovered.Conclusion. Our study demonstrated the potential of Mg/Al-LDH as an effective adjuvant for pertussis vaccine, which induced comparable antibody response and had a better safety compared with commercial AH adjuvant.
Collapse
Affiliation(s)
- Dongdong Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, No. 2699 Qianjin Street, Changchun 130012, People's Republic of China
| | - Mengjie Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, No. 2699 Qianjin Street, Changchun 130012, People's Republic of China
| | - Gaotian Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, No. 2699 Qianjin Street, Changchun 130012, People's Republic of China
| | - Yu Zheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, No. 2699 Qianjin Street, Changchun 130012, People's Republic of China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, No. 2699 Qianjin Street, Changchun 130012, People's Republic of China
| | - Dandan Xia
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, No. 26, Huatuo Street, Benxi 117004, People's Republic of China
| | - Shaoning Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, No. 26, Huatuo Street, Benxi 117004, People's Republic of China
| | - Yan Chen
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, No. 2699 Qianjin Street, Changchun 130012, People's Republic of China
| |
Collapse
|
13
|
Wang Z, Liu Y, Peng L, Till B, Liao Y, Yuan S, Yan X, Chen L, Fu Q, Qin Z. Role of fibrosarcoma-induced CD11b + myeloid cells and tumor necrosis factor-α in B cell responses. Oncogene 2022; 41:1434-1444. [PMID: 35034094 DOI: 10.1038/s41388-022-02187-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/20/2021] [Accepted: 01/10/2022] [Indexed: 11/09/2022]
Abstract
The role of B cells in the anti-tumor immune response remains controversial. An increase in the number of B cells in the peripheral blood of some tumor patients has been associated with poor immunotherapy efficacy. However, the mechanism leading to the generation of these cells is not well-described. Using a fibrosarcoma model, we show that intraperitoneal administration of a xenogeneic antigen in tumor-bearing mice evokes large increases in antigen-specific serum immunoglobulin formation compared to tumor-naïve mice. An inability of tumor-bearing mice to induce enhanced antibody production after myeloid cell depletion suggests the antibody responses are CD11b+ myeloid cell-dependent. In vitro, CD11b+ myeloid cells promoted B cell proliferation, activation, and survival. High levels of tumor necrosis factor (TNF)-α were produced by CD11b+ cells, and TNF-α blockade inhibited B cell responses. CD11b+ cells appear to be important promoters of B cell responses and targeting B cells may increase the efficacy of immunotherapy in tumor-bearing hosts.
Collapse
Affiliation(s)
- Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| | - Yuqing Liu
- Department of Oncology, Third Affiliated Hospital of Xinxiang Medical College, Xinxiang, China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Brian Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yuwei Liao
- Yangjiang Key Laboratory of Respiratory Diseases, Yangjiang People's Hospital, Yangjiang, China
| | - Shumin Yuan
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiang Yan
- Medical Oncology Department, Chinese PLA General Hospital, Beijing, China
| | - Lin Chen
- GZMU-GIBH School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Qiang Fu
- Department of General Surgery, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Shi Y. PLAN B for immunotherapy: Promoting and leveraging anti-tumor B cell immunity. J Control Release 2021; 339:156-163. [PMID: 34563591 DOI: 10.1016/j.jconrel.2021.09.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 12/19/2022]
Abstract
Current immuno-oncology primarily focuses on adaptive cellular immunity mediated by T lymphocytes. The other important lymphocytes, B cells, are largely ignored in cancer immunotherapy. B cells are generally considered to be responsible for humoral immune response to viral and bacterial infections. The role of B cells in cancer immunity has long been under debate. Recently, increasing evidence from both preclinical and clinical research has shown that B cells can also induce potent anti-cancer immunity, via humoral and cellular immune responses. Yet it is unclear how to efficiently integrate B cell immunity in cancer immunotherapy. In the current perspective, anti-tumor immunity of B cells is discussed regarding antibody production, antigen presentation, cytokine release and contribution to intratumoral tertiary lymphoid structures. Afterwards, immunosuppressive regulatory phenotypes of B cells are summarized. Furthermore, strategies to activate and modulate B cells using nanomedicines and biomaterials are discussed. This article provides a unique perspective on "PLAN B" (promoting and leveraging anti-tumor B cell immunity) using nanomedicines and biomaterials for cancer immunotherapy. This is envisaged to form a new research direction with the potential to reach the next breakthrough in immunotherapy.
Collapse
Affiliation(s)
- Yang Shi
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen 52074, Germany.
| |
Collapse
|
15
|
Zhou M, Dascani P, Ding C, Kos JT, Tieri D, Lin X, Caster D, Powell D, Wen C, Watson CT, Yan J. Integrin CD11b Negatively Regulates B Cell Receptor Signaling to Shape Humoral Response during Immunization and Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2021; 207:1785-1797. [PMID: 34470858 DOI: 10.4049/jimmunol.2100070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/29/2021] [Indexed: 11/19/2022]
Abstract
Our previous work has revealed the ability of CD11b to regulate BCR signaling and control autoimmune disease in mice. However, how CD11b regulates the immune response under normal conditions remains unknown. Through the use of a CD11b knockout model on a nonautoimmune background, we demonstrated that CD11b-deficient mice have an elevated Ag-specific humoral response on immunization. Deletion of CD11b resulted in elevated low-affinity and high-affinity IgG Ab and increases in Ag-specific germinal center B cells and plasma cells (PCs). Examination of BCR signaling in CD11b-deficient mice revealed defects in association of negative regulators pLyn and CD22 with the BCR, but increases in colocalizations between positive regulator pSyk and BCR after stimulation. Using a CD11b-reporter mouse model, we identified multiple novel CD11b-expressing B cell subsets that are dynamically altered during immunization. Subsequent experiments using a cell-specific CD11b deletion model revealed this effect to be B cell intrinsic and not altered by myeloid cell CD11b expression. Importantly, CD11b expression on PCs also impacts on BCR repertoire selection and diversity in autoimmunity. These studies describe a novel role for CD11b in regulation of the healthy humoral response and autoimmunity, and reveal previously unknown populations of CD11b-expressing B cell subsets, suggesting a complex function for CD11b in B cells during development and activation.
Collapse
Affiliation(s)
- Mingqian Zhou
- Division of Immunotherapy, The Hiram C. Polk, Jr. Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY.,College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Paul Dascani
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY
| | - Chuanlin Ding
- Division of Immunotherapy, The Hiram C. Polk, Jr. Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY
| | - Justin T Kos
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY; and
| | - David Tieri
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY; and
| | - Xiaoying Lin
- Division of Immunotherapy, The Hiram C. Polk, Jr. Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY.,College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dawn Caster
- Department of Medicine, University of Louisville, Louisville, KY
| | - David Powell
- Department of Medicine, University of Louisville, Louisville, KY
| | - Chengping Wen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Corey T Watson
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY; and
| | - Jun Yan
- Division of Immunotherapy, The Hiram C. Polk, Jr. Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY; .,Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY
| |
Collapse
|
16
|
Zhu J, Ananthaswamy N, Jain S, Batra H, Tang WC, Lewry DA, Richards ML, David SA, Kilgore PB, Sha J, Drelich A, Tseng CTK, Chopra AK, Rao VB. A universal bacteriophage T4 nanoparticle platform to design multiplex SARS-CoV-2 vaccine candidates by CRISPR engineering. SCIENCE ADVANCES 2021; 7:eabh1547. [PMID: 34516878 PMCID: PMC8442874 DOI: 10.1126/sciadv.abh1547] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/16/2021] [Indexed: 06/02/2023]
Abstract
A “universal” platform that can rapidly generate multiplex vaccine candidates is critically needed to control pandemics. Using the severe acute respiratory syndrome coronavirus 2 as a model, we have developed such a platform by CRISPR engineering of bacteriophage T4. A pipeline of vaccine candidates was engineered by incorporating various viral components into appropriate compartments of phage nanoparticle structure. These include expressible spike genes in genome, spike and envelope epitopes as surface decorations, and nucleocapsid proteins in packaged core. Phage decorated with spike trimers was found to be the most potent vaccine candidate in animal models. Without any adjuvant, this vaccine stimulated robust immune responses, both T helper cell 1 (TH1) and TH2 immunoglobulin G subclasses, blocked virus-receptor interactions, neutralized viral infection, and conferred complete protection against viral challenge. This new nanovaccine design framework might allow the rapid deployment of effective adjuvant-free phage-based vaccines against any emerging pathogen in the future.
Collapse
Affiliation(s)
- Jingen Zhu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Neeti Ananthaswamy
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Swati Jain
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Himanshu Batra
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Wei-Chun Tang
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | | | | | | | - Paul B. Kilgore
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jian Sha
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Aleksandra Drelich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Chien-Te K. Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ashok K. Chopra
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Venigalla B. Rao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
17
|
Combination of conserved recombinant proteins (NP & 3M2e) formulated with Alum protected BALB/c mice against influenza A/PR8/H1N1 virus challenge. Biotechnol Lett 2021; 43:2137-2147. [PMID: 34491470 DOI: 10.1007/s10529-021-03174-2] [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: 02/07/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Influenza is one of the most important agents of pandemic outbreak causing substantial morbidity and mortality. Vaccination strategies of influenza must be adapted annually due to constant antigenic changes in various strains. Therefore, the present study was conducted to evaluate protective immunity of the conserved influenza proteins. METHODS For this purpose, three tandem repeats of M2e (3M2e) and NP were separately expressed in E. coli and were purified using column chromatography. Female Balb/c mice were injected intradermally with a combination of the purified 3M2e and NP alone or formulated with Alum (AlOH3) adjuvant in three doses. The mice were challenged by intranasal administration of H1N1 (A/PR/8/34) 2 weeks after the last vaccination. RESULTS The results demonstrated that recombinant NP and M2e proteins are immunogenic and could efficiently elicit immune responses in mice compared to non-immunized mice. The combination of 3M2e and NP supplemented with Alum stimulated both NP and M2e-specific antibodies, which were higher than those stimulated by each single antigen plus Alum. In addition, the secretion of IFN-γ and IL-4 as well as the induction of lymphocyte proliferation in mice received the mixture of these proteins with Alum was considerably higher than other groups. Moreover, the highest survival rate (86%) with the least body weight change was observed in the mice immunized with 3M2e and NP supplemented with Alum followed by the mice received NP supplemented with Alum (71%). CONCLUSION Accordingly, this regimen can be considered as an attractive candidate for global vaccination against influenza.
Collapse
|
18
|
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: 18] [Impact Index Per Article: 4.5] [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
|
19
|
Shardlow E, Linhart C, Connor S, Softely E, Exley C. The measurement and full statistical analysis including Bayesian methods of the aluminium content of infant vaccines. J Trace Elem Med Biol 2021; 66:126762. [PMID: 33887692 DOI: 10.1016/j.jtemb.2021.126762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Aluminium salts are the most common adjuvants in infant vaccines. The aluminium content of a vaccine is provided by the manufacturer and is indicated on the patient information leaflet. There is no independent verification, for example by the European Medicines Agency, of the aluminium content of infant vaccines. METHODS We have measured the aluminium content of thirteen infant vaccines using microwave-assisted acid and peroxide digestion followed by transversely heated graphite furnace atomic absorption spectrometry. Our data are compared with manufacturer's data using full statistical analyses including Bayesian methods. RESULTS We found that only three vaccines contained the amount of aluminium indicated by the manufacturer. Six vaccines contained a statistically significant (P < 0.05) greater quantity while four vaccines contained a statistically significant (P < 0.05) lower quantity. The range of content for any single vaccine varied considerably, for example, from 0.172 to 0.602 mg/vaccine for Havrix. CONCLUSIONS The data have raised specific questions about the significance of the aluminium content of vaccines and identified areas of extremely limited information. Since aluminium is a known toxin in humans and specifically a neurotoxin, its content in vaccines should be accurate and independently monitored to ensure both efficacy and safety.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| | - Caroline Linhart
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| | - Sameerah Connor
- Life Sciences, The Huxley Building, Keele University, Staffordshire, United Kingdom
| | - Erin Softely
- Life Sciences, The Huxley Building, Keele University, Staffordshire, United Kingdom
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom.
| |
Collapse
|
20
|
Folci M, Ramponi G, Arcari I, Zumbo A, Brunetta E. Eosinophils as Major Player in Type 2 Inflammation: Autoimmunity and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:197-219. [PMID: 34031864 DOI: 10.1007/5584_2021_640] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Eosinophils are a subset of differentiated granulocytes which circulate in peripheral blood and home in several body tissues. Along with their traditional relevance in helminth immunity and allergy, eosinophils have been progressively attributed important roles in a number of homeostatic and pathologic situations. This review aims at summarizing available evidence about eosinophils functions in homeostasis, infections, allergic and autoimmune disorders, and solid and hematological cancers.Their structural and biological features have been described, along with their physiological behavior. This includes their chemokines, cytokines, granular contents, and extracellular traps. Besides, pathogenic- and eosinophilic-mediated disorders have also been addressed, with the aim of highlighting their role in Th2-driven inflammation. In allergy, eosinophils are implicated in the pathogenesis of atopic dermatitis, allergic rhinitis, and asthma. They are also fundamentally involved in autoimmune disorders such as eosinophilic esophagitis, eosinophilic gastroenteritis, acute and chronic eosinophilic pneumonia, and eosinophilic granulomatosis with polyangiitis. In infections, eosinophils are involved in protection not only from parasites but also from fungi, viruses, and bacteria. In solid cancers, local eosinophilic infiltration is variably associated with an improved or worsened prognosis, depending on the histotype. In hematologic neoplasms, eosinophilia can be the consequence of a dysregulated cytokine production or the result of mutations affecting the myeloid lineage.Recent experimental evidence was thoroughly reviewed, with findings which elicit a complex role for eosinophils, in a tight balance between host defense and tissue damage. Eventually, emerging evidence about eosinophils in COVID-19 infection was also discussed.
Collapse
Affiliation(s)
- Marco Folci
- Humanitas Clinical and Research Center - IRCCS, Milan, Italy. .,Department of Biomedical Sciences, Humanitas University, Milan, Italy.
| | - Giacomo Ramponi
- Humanitas Clinical and Research Center - IRCCS, Milan, Italy
| | - Ivan Arcari
- Humanitas Clinical and Research Center - IRCCS, Milan, Italy
| | - Aurora Zumbo
- Humanitas Clinical and Research Center - IRCCS, Milan, Italy
| | - Enrico Brunetta
- Humanitas Clinical and Research Center - IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
21
|
Du L, Chang H, Xu W, Zhang X, Yin L. Elevated chemokines and cytokines for eosinophils in neuromyelitis optica spectrum disorders. Mult Scler Relat Disord 2021; 52:102940. [PMID: 33930716 DOI: 10.1016/j.msard.2021.102940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/27/2021] [Accepted: 04/02/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Eosinophil infiltration is one of the distinctive features in neuromyelitis optica spectrum disorders (NMOSD) but not in other demyelinating diseases including multiple sclerosis (MS). Eosinophils express the chemokine receptor CCR3, which is activated by eotaxins (eotaxin-1, -2, and -3) and monocyte chemoattractant protein (MCP)-4. We aimed to investigate the role of MCPs (MCP-1, -2, -3, and -4) and eotaxins in the acute phase of NMOSD. METHODS Levels of serum and cerebrospinal fluid (CSF) eotaxins, MCPs, interleukin (IL)-5, tumor necrosis factor (TNF)-α, granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL-6 were measured using the cytokine multiplex assay from 26 patients with NMOSD (13 with immunotherapy, 13 without immunotherapy), 9 patients with MS, and 9 patients with other noninflammatory neurological diseases (OND). Glial fibrillary acidic protein was assessed using ELISA. RESULTS Serum MCP-1 and CSF MCP-2 levels were significantly higher in patients with NMOSD than in OND. Moreover, serum MCP-4 and CSF eotaxin-2 and -3 levels were significantly higher in NMOSD patients compared to MS and OND. Serum MCP-1, -4 and CSF eotaxin-2, -3 levels were significantly correlated with the Expanded Disability Status Scale in NMOSD. TNF-α and GM-CSF, which stimulate the above chemokines, were higher in patients with NMOSD than those in OND. Moreover, serum MCP-1 and -4 were significantly increased by IL-5 and GM-CSF stimulation, but not by TNF-α and IL-6. Only CSF eotaxin-2 was significantly increased by GM-CSF. There were no significant differences in serum MCP-1 and -4 levels between NMOSD patients with and without immunotherapy. CONCLUSION These findings suggest that the elevated serum MCP-1, -4 and CSF eotaxin-2, -3 may be a key step in eosinophil recruitment in the acute phase of NMOSD.
Collapse
Affiliation(s)
- Li Du
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Haoxiao Chang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wangshu Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xinghu Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Linlin Yin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
22
|
Du Y, Xu Y, Feng J, Hu L, Zhang Y, Zhang B, Guo W, Mai R, Chen L, Fang J, Zhang H, Peng T. Intranasal administration of a recombinant RBD vaccine induced protective immunity against SARS-CoV-2 in mouse. Vaccine 2021; 39:2280-2287. [PMID: 33731271 PMCID: PMC7934688 DOI: 10.1016/j.vaccine.2021.03.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/06/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
The emergence of the global Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) pandemic underscores the importance of the rapid development of a non-invasive vaccine that can be easily administered. A vaccine administered by nasal delivery is endowed with such characteristics against respiratory viruses. In this study, we generated a recombinant SARS-CoV-2 receptor-binding domain (RBD)-based subunit vaccine. Mice were immunized via intranasal inoculation, microneedle-intradermal injection, or intramuscular injection, after which the RBD-specific immune responses were compared. Results showed that when administrated intranasally, the vaccine elicited a robust systemic humoral immunity with high titers of IgG antibodies and neutralizing antibodies as well as a significant mucosal immunity. Besides, antigen-specific T cell responses were also analyzed. These results indicated that the non-invasive intranasal administration should be explored for the future SARS-CoV-2 vaccine design.
Collapse
Affiliation(s)
- Yingying Du
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuhua Xu
- Guangdong South China Vaccine, Guangzhou, China
| | - Jin Feng
- Guangdong South China Vaccine, Guangzhou, China
| | - Longbo Hu
- Sino-French Hoffmann Institute of Immunology, State Key Laboratory of Respiratory Disease, College of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yanan Zhang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Bo Zhang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Weili Guo
- Guangdong South China Vaccine, Guangzhou, China
| | - Runming Mai
- Guangdong South China Vaccine, Guangzhou, China
| | - Liyun Chen
- Guangdong South China Vaccine, Guangzhou, China
| | - Jianmin Fang
- Sino-French Hoffmann Institute of Immunology, State Key Laboratory of Respiratory Disease, College of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Tao Peng
- Guangdong South China Vaccine, Guangzhou, China; Sino-French Hoffmann Institute of Immunology, State Key Laboratory of Respiratory Disease, College of Basic Medical Science, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
23
|
Rincon JC, Hawkins RB, Hollen M, Nacionales DC, Ungaro R, Efron PA, Moldawer LL, Larson SD. Aluminum Adjuvant Improves Survival Via NLRP3 Inflammasome and Myeloid Non-Granulocytic Cells in a Murine Model of Neonatal Sepsis. Shock 2021; 55:274-282. [PMID: 32769820 PMCID: PMC8025597 DOI: 10.1097/shk.0000000000001623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT Neonatal sepsis leads to significant morbidity and mortality with the highest risk of death occurring in preterm (<37 weeks) and low birth weight (<2,500 g) infants. The neonatal immune system is developmentally immature with well-described defects in innate and adaptive immune responses. Immune adjuvants used to enhance the vaccine response have emerged as potential therapeutic options, stimulating non-specific immunity and preventing sepsis mortality. Aluminum salts ("alum") have been used as immune adjuvants for over a century, but their mechanism of action remains poorly understood. This study aims to identify potential mechanisms by which pretreatment with alum induces host protective immunity to polymicrobial sepsis in neonatal mice. Utilizing genetic and cell-depletion studies, we demonstrate here that the prophylactic administration of aluminum adjuvants in neonatal mice improves sepsis survival via activation of the nucleotide oligomerization domain-like receptor family, pyrin-domain-containing 3 inflammasome and dendritic cell activation. Furthermore, this beneficial effect is dependent on myeloid, non-granulocytic Gr1-positive cells, and MyD88-signaling pathway activation. These findings suggest a promising therapeutic role for aluminum-based vaccine adjuvants to prevent development of neonatal sepsis and improve mortality in this highly vulnerable population.
Collapse
Affiliation(s)
- Jaimar C Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhu J, Ananthaswamy N, Jain S, Batra H, Tang WC, Lewry DA, Richards ML, David SA, Kilgore PB, Sha J, Drelich A, Tseng CTK, Chopra AK, Rao VB. A Universal Bacteriophage T4 Nanoparticle Platform to Design Multiplex SARS-CoV-2 Vaccine Candidates by CRISPR Engineering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.19.427310. [PMID: 33501450 PMCID: PMC7836120 DOI: 10.1101/2021.01.19.427310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A "universal" vaccine design platform that can rapidly generate multiplex vaccine candidates is critically needed to control future pandemics. Here, using SARS-CoV-2 pandemic virus as a model, we have developed such a platform by CRISPR engineering of bacteriophage T4. A pipeline of vaccine candidates were engineered by incorporating various viral components into appropriate compartments of phage nanoparticle structure. These include: expressible spike genes in genome, spike and envelope epitopes as surface decorations, and nucleocapsid proteins in packaged core. Phage decorated with spike trimers is found to be the most potent vaccine candidate in mouse and rabbit models. Without any adjuvant, this vaccine stimulated robust immune responses, both T H 1 and T H 2 IgG subclasses, blocked virus-receptor interactions, neutralized viral infection, and conferred complete protection against viral challenge. This new type of nanovaccine design framework might allow rapid deployment of effective phage-based vaccines against any emerging pathogen in the future.
Collapse
|
25
|
Wu P, Zhang Y, Yin X, He Y, Zhang Q, Chen C. Layered double hydroxide nanoparticles as an adjuvant for inactivated foot-and-mouth disease vaccine in pigs. BMC Vet Res 2020; 16:474. [PMID: 33276787 PMCID: PMC7716589 DOI: 10.1186/s12917-020-02689-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 11/23/2020] [Indexed: 01/19/2023] Open
Abstract
Background Foot-and-mouth disease (FMD) is a highly transmissible disease that leads to vast economic losses in many countries. Prevention using inactivated vaccines is one effective measure used to control FMD. Unfortunately, inactivated FMD vaccines provide only short-term protection and require a cold-chain system. In recent years, many studies have shown that layered double metal hydroxides (LDHs) carrying antigens can be used to strongly induce immune responses. In this study, LDH nanoparticles (NPs) were prepared by hydrothermal synthesis. LDH particle size, electric potential, and morphology were measured and observed. The adsorption capacity of LDH NPs to FMDV was tested. The effects of LDH as an adjuvant on inactivated FMDV vaccines were further evaluated and compared with commercial FMDV Montanide ISA-206 in BALB/C female mice and Yorkshire pigs. Results LDH NPs were successfully prepared with a uniform particle size of ~ 87.21 nm, regular edges, a loose hexagonal shape and positive zeta charge of 32 mV. The maximum absorption concentration was 0.16–0.31 μg FMDV/μg LDH. In the mouse experiment, antibody levels in group LDH + FMDV were significantly higher compared to group saline + FMDV (P < 0.01) from days 42–98 and were significantly higher to group ISA-206 + FMDV on day 56 post-immunization (P < 0.05). After day 14 post-immunization, IFN-γ content was significantly increased (P < 0.05). In the pig experiment, antibody levels in both the ISA-206 + FMDV and LDH + FMDV were positive and were significantly higher compared with the PBS group on day 7 (P < 0.005). Antibody levels in 90% pigs were positive on day 56 in the LDH group. The neutralizing antibody levels in the LDH and ISA-206 groups were significantly higher from days 7–28 compared to the PBS control group (P < 0.05). Thus, LDH NPs were effective at inducing an immune response against FMDV. Conclusions LDHs with a loose hexagonal shape and a positive charge were prepared and evaluated as adjuvant for FMD vaccine. It was demonstrated that LDHs can induce immune responses in mice and pigs. In addition, the LDHs produced antibodies continuously which may indicate a slow-release effect. The study shows that LDHs may act as a potentially useful FMDV adjuvant.
Collapse
Affiliation(s)
- Peng Wu
- College of Animal Science and Technology, Shihezi University, Xinjiang, China.,College of Life Technology, Shihezi University, Xinjiang, China
| | - Yunfeng Zhang
- College of Animal Science and Technology, Shihezi University, Xinjiang, China.,State Key Laboratory of Sheep Genetic Improvement and Healthy Production/ Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | - Xinyue Yin
- College of Animal Science and Technology, Shihezi University, Xinjiang, China
| | - Yanhua He
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production/ Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | - Qian Zhang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production/ Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Xinjiang, China.
| |
Collapse
|
26
|
Dong Y, Dai T, Wei Y, Zhang L, Zheng M, Zhou F. A systematic review of SARS-CoV-2 vaccine candidates. Signal Transduct Target Ther 2020; 5:237. [PMID: 33051445 PMCID: PMC7551521 DOI: 10.1038/s41392-020-00352-y] [Citation(s) in RCA: 377] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/06/2020] [Accepted: 09/27/2020] [Indexed: 01/18/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging virus that is highly pathogenic and has caused the recent worldwide pandemic officially named coronavirus disease (COVID-19). Currently, considerable efforts have been put into developing effective and safe drugs and vaccines against SARS-CoV-2. Vaccines, such as inactivated vaccines, nucleic acid-based vaccines, and vector vaccines, have already entered clinical trials. In this review, we provide an overview of the experimental and clinical data obtained from recent SARS-CoV-2 vaccines trials, and highlight certain potential safety issues that require consideration when developing vaccines. Furthermore, we summarize several strategies utilized in the development of vaccines against other infectious viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), with the aim of aiding in the design of effective therapeutic approaches against SARS-CoV-2.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2
- Antibodies, Viral/biosynthesis
- Betacoronavirus/drug effects
- Betacoronavirus/immunology
- Betacoronavirus/pathogenicity
- COVID-19
- COVID-19 Vaccines
- Clinical Trials as Topic
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/virology
- Gene Expression Regulation/drug effects
- Humans
- Immunity, Innate/drug effects
- Immunization Schedule
- Immunogenicity, Vaccine
- Middle East Respiratory Syndrome Coronavirus/drug effects
- Middle East Respiratory Syndrome Coronavirus/immunology
- Middle East Respiratory Syndrome Coronavirus/pathogenicity
- Pandemics/prevention & control
- Patient Safety
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- Protein Binding
- Receptors, Virus/antagonists & inhibitors
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Severe acute respiratory syndrome-related coronavirus/drug effects
- Severe acute respiratory syndrome-related coronavirus/immunology
- Severe acute respiratory syndrome-related coronavirus/pathogenicity
- SARS-CoV-2
- Severe Acute Respiratory Syndrome/immunology
- Severe Acute Respiratory Syndrome/prevention & control
- Severe Acute Respiratory Syndrome/virology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/metabolism
- Vaccines, Attenuated
- Vaccines, DNA
- Vaccines, Subunit
- Vaccines, Virus-Like Particle
- Viral Vaccines/administration & dosage
- Viral Vaccines/biosynthesis
Collapse
Affiliation(s)
- Yetian Dong
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Hangzhou, 310058, China
| | - Tong Dai
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China
| | - Yujun Wei
- Anhui Anlong Gene Technology Co., Ltd, Hefei, 230041, China
| | - Long Zhang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Hangzhou, 310058, China
| | - Min Zheng
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
27
|
Alqahtani MS, Kazi M, Ahmad MZ, Syed R, Alsenaidy MA, Albraiki SA. Lignin nanoparticles as a promising vaccine adjuvant and delivery system for ovalbumin. Int J Biol Macromol 2020; 163:1314-1322. [PMID: 32645499 DOI: 10.1016/j.ijbiomac.2020.07.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 10/23/2022]
Abstract
Vaccination is the most effective strategy of preventing and treating infectious diseases and the most significant issue in the development of potent vaccines is the sufficient immunogenicity and safety of vaccines. The main goal of the present study is to develop a potent and safe vaccine adjuvant that can also stabilize antigen formulations during preparation and storage. In this study, the model antigen ovalbumin (OVA) was encapsulated in polymeric nanoparticles based on lignin (OVA-LNPs). The nanoparticles had a particle size of 216 nm and a low polydispersity index. The nanoparticles were negatively charged (-26.7 mV) with high encapsulation efficiency 81.6% of OVA antigen. In vitro studies of the nanoparticles were tested against dendritic cells (DCs), specialized antigen-presenting cells (APCs). The results showed no cytotoxic effect from LNPs and a significantly higher percentage of dendritic cells have taken up the antigen when encapsulated inside LNPs in contrast to free OVA. The nanoparticle was administered intradermally to BALB/c mice and the resulting time-dependent systemic immune responses towards OVA were assessed by measuring the OVA-specific IgG titers using an enzyme-linked immunosorbent assay (ELISA). In vivo immunization with OVA-LNPs induced a stronger IgG antibody response than that induced by free OVA or alum adjuvanted OVA. Enhanced immunization by OVA-LNPs was attributed to the observed efficient uptake of the antigen by dendritic cells. These findings demonstrate that LNPs are promising to be used as vaccine adjuvant and delivery system for the induction of long-term immune responses.
Collapse
Affiliation(s)
- Mohammed S Alqahtani
- Department of Pharmaceutics, College of Pharmacy, PO Box 2457, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, PO Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Muhammad Z Ahmad
- Department of Pharmacognosy, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia
| | - Rabbani Syed
- Department of Pharmaceutics, College of Pharmacy, PO Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad A Alsenaidy
- Department of Pharmaceutics, College of Pharmacy, PO Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Salem A Albraiki
- Department of Pharmaceutics, College of Pharmacy, PO Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
28
|
Lacy P. Gr1 makes an unexpected cameo appearance in eosinophils. J Leukoc Biol 2020; 107:363-365. [PMID: 31985092 DOI: 10.1002/jlb.2ce1119-473r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/25/2019] [Accepted: 01/13/2020] [Indexed: 01/10/2023] Open
Abstract
Discussion of the neutrophil marker Ly6G/Gr1, which is readily detectable on mouse eosinophils, and is expressed in higher levels in the presence of IL-5.
Collapse
Affiliation(s)
- Paige Lacy
- Alberta Respiratory Centre, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
29
|
Shardlow E, Mold M, Exley C. The interaction of aluminium-based adjuvants with THP-1 macrophages in vitro: Implications for cellular survival and systemic translocation. J Inorg Biochem 2019; 203:110915. [PMID: 31751817 DOI: 10.1016/j.jinorgbio.2019.110915] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023]
Abstract
Within clinical vaccinations, recombinant antigens are routinely entrapped inside or adsorbed onto the surface of aluminium salts in order to increase their immunological potency in vivo. The efficacy of these immunisations is highly dependent upon the recognition and uptake of these complexes by professional phagocytes and their subsequent delivery to the draining lymph nodes for further immunological processing. While monocytes have been shown to internalise aluminium adjuvants and their adsorbates, the role of macrophages in this respect has not been fully established. Furthermore, this study explored the interaction of THP-1 macrophages with aluminium-based adjuvants (ABAs) and how this relationship influenced the survival of such cells in vitro. THP-1 macrophages were exposed to low concentrations of ABAs (1.7 μg/mL Al) for a maximum of seven days. ABA uptake was determined using lumogallion staining and cell viability by both DAPI (4',6-diamidino-2-phenylindole) staining and LDH (lactate dehydrogenase) assay. Evidence of ABA particle loading was identified within cells at early junctures following treatment and appeared to be quite prolific (>90% cells positive for Al signal after 24 h). Total sample viability (% LDH release) in treated samples was predominantly similar to untreated cells and low levels of cellular death were consistently observed in populations positive for Al uptake. It can thus be concluded that aluminium salts can persist for some time within the intracellular environment of these cells without adversely affecting their viability. These results imply that macrophages may play a role in the systemic translocation of ABAs once administered in the form of an inoculation.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK
| | - Matthew Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK.
| |
Collapse
|
30
|
Limkar AR, Mai E, Sek AC, Percopo CM, Rosenberg HF. Frontline Science: Cytokine-mediated developmental phenotype of mouse eosinophils: IL-5-associated expression of the Ly6G/Gr1 surface Ag. J Leukoc Biol 2019; 107:367-377. [PMID: 31674692 DOI: 10.1002/jlb.1hi1019-116rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/09/2019] [Accepted: 10/12/2019] [Indexed: 01/21/2023] Open
Abstract
Eosinophils have broad and extensive immunomodulatory capacity; recent studies have focused on the roles of distinct eosinophil subsets in specific tissue microenvironments. Ly6G is a GPI-linked leukocyte surface Ag understood primarily as a marker of mouse neutrophils, although its full function is not known. Here, we show that Ly6G/Gr1, detected by mAbs 1A8 (anti-Ly6G) and RB6-8C5 (anti-Gr1), is detected prominently on a significant fraction of eosinophils from mouse bone marrow and bone marrow-derived culture, with fractions expressing this Ag increasing in IL-5-enriched microenvironments. Among our findings, we identified SiglecF+ Gr1+ eosinophils in bone marrow from naïve, allergen-challenged and IL-5 transgenic mice; SiglecF+ Gr1+ eosinophils were also prominent ex vivo in bone marrow-derived eosinophils (bmEos) in IL-5-enriched culture. Reducing the IL-5 concentration 20-fold had no impact on the rate of generation of SiglecF+ bmEos but did result in a marked increase in the Gr1- fraction (from 17.4 ± 2% to 30 ± 2.3%, ***P < 0.005). Reducing the IL-5 concentration also enhanced chemotaxis; SiglecF+ Gr1- bmEos were considerably more responsive to eotaxin-1 than were their SiglecF+ Gr1+ counterparts. These results suggest that (i) IL-5 regulates the expression of Ly6G/Gr1, either directly or indirectly, in cells of the eosinophil lineage, (ii) eosinophils generated in response to high concentrations of IL-5 can be distinguished from those generated under homeostatic conditions by expression of the Ly6G/Gr1 cell surface Ag, and (iii) expression of Ly6G/Gr1 may have an impact on function, directly or indirectly, including the potential to undergo chemotaxis in response to eotaxin-1.
Collapse
Affiliation(s)
- Ajinkya R Limkar
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric Mai
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Albert C Sek
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Caroline M Percopo
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Shardlow E, Exley C. The size of micro-crystalline tyrosine (MCT®) influences its recognition and uptake by THP-1 macrophages in vitro. RSC Adv 2019; 9:24505-24518. [PMID: 35527856 PMCID: PMC9069726 DOI: 10.1039/c9ra03831k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/26/2019] [Indexed: 11/21/2022] Open
Abstract
The physicochemical hallmarks of particulate immunopotentiators play a pivotal role with regards to their adjuvanticity in vivo. These properties have not been fully characterised in the case of MCT®, an amino acid-based adjuvant used as an alternative to aluminium salts in subcutaneous allergy immunotherapy (SCIT). This study presents a full characterisation of MCT® and in a preliminary capacity reveals how parameters, specifically particle size, might influence the recognition of MCT® by antigen presenting cells (APCs) in vitro. Light microscopic analysis demonstrated that MCT® was composed of highly crystalline needles, the majority of which exceeded 10 μm in length under physiological conditions (median size – 20.8 μm). While the substantial length of crystals presented a significant barrier to cellular recognition and uptake, isolated incidences of perpendicular recognition were observed owing to the smaller comparative width of crystallites (median size – 2.8 μm). This appeared to allow a small proportion of material to be ingested both fully and partially by THP-1 macrophages, although further studies are required to unequivocally confirm this observation. Preferential recognition of needle tips also favoured the direct presentation of antigen to immune cells as proteinaceous adsorption appeared to be isolated to these regions. Furthermore, the data herein provide valuable insights into the mechanisms surrounding how this adjuvant potentiates an immunological response following administration. The large size of MCT® crystallites partially stymies their recognition and uptake by THP-1 macrophages in vitro.![]()
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University Keele Staffordshire ST5 5BG UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University Keele Staffordshire ST5 5BG UK
| |
Collapse
|
32
|
Ribechini E, Eckert I, Beilhack A, Du Plessis N, Walzl G, Schleicher U, Ritter U, Lutz MB. Heat-killed Mycobacterium tuberculosis prime-boost vaccination induces myeloid-derived suppressor cells with spleen dendritic cell-killing capability. JCI Insight 2019; 5:128664. [PMID: 31162143 DOI: 10.1172/jci.insight.128664] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis patients and mice infected with live Mycobacterium tuberculosis (Mtb) accumulate high numbers of myeloid-derived suppressor cells (MDSCs). Here, we hypothesized that also dead Mtb vaccines may induce MDSCs that could impair the efficacy of vaccination. We found that repeated injections of Mtb vaccines (heat-killed Mtb in Incomplete Freund's Adjuvant, like Montanide) but not single or control vaccines without Mtb strongly expanded CD11b+ myeloid cells in the spleen, that suppressed T cell proliferation and killing ex vivo. Dead Mtb vaccination induced the generation of CD11b+ Ly-6Chigh CD115+ iNOS/Nos2+ monocytic MDSCs (M-MDSCs) upon application of inflammatory or microbial activation signals. In vivo these M-MDSCs positioned strategically in the spleen by infiltrating the splenic bridging channels and white pulp areas. Notably, within 6 to 24 hours in a Nos2-dependent fashion they produced NO to rapidly kill conventional and plasmacytoid dendritic cells (cDCs, pDCs) while, surprisingly, sparing T cells in vivo. Thus, we demonstrate that Mtb vaccine induced M-MDSCs to not directly suppress T cell in vivo but, instead, M-MDSCs directly target DC subpopulations thereby indirectly suppressing effector T cell responses. Collectively, we demonstrate that Mtb booster vaccines induce M-MDSCs in the spleen that can be activated to kill DCs cautioning to thoroughly investigate MDSC formation in individuals after Mtb vaccination in clinical trials.
Collapse
Affiliation(s)
- Eliana Ribechini
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Ina Eckert
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Nelita Du Plessis
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology - National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology - National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ulrike Schleicher
- Microbiology Institute, Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Uwe Ritter
- RCI Regensburg Center for Interventional Immunology, Institute of Immunology, University Medical Center Regensburg, University of Regensburg, Regensburg, Germany
| | - Manfred B Lutz
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
33
|
Sarkar I, Garg R, van Drunen Littel-van den Hurk S. Selection of adjuvants for vaccines targeting specific pathogens. Expert Rev Vaccines 2019; 18:505-521. [PMID: 31009255 PMCID: PMC7103699 DOI: 10.1080/14760584.2019.1604231] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Adjuvants form an integral component in most of the inactivated and subunit vaccine formulations. Careful and proper selection of adjuvants helps in promoting appropriate immune responses against target pathogens at both innate and adaptive levels such that protective immunity can be elicited. Areas covered: Herein, we describe the recent progress in our understanding of the mode of action of adjuvants that are licensed for use in human vaccines or in clinical or pre-clinical stages at both innate and adaptive levels. Different pathogens have distinct characteristics, which require the host to mount an appropriate immune response against them. Adjuvants can be selected to elicit a tailor-made immune response to specific pathogens based on their unique properties. Identification of biomarkers of adjuvanticity for several candidate vaccines using omics-based technologies can unravel the mechanism of action of modern and experimental adjuvants. Expert opinion: Adjuvant technology has been revolutionized over the last two decades. In-depth understanding of the role of adjuvants in activating the innate immune system, combined with systems vaccinology approaches, have led to the development of next-generation, novel adjuvants that can be used in vaccines against challenging pathogens and in specific target populations.
Collapse
Affiliation(s)
- Indranil Sarkar
- a VIDO-InterVac , University of Saskatchewan , Saskatoon , Canada.,b Microbiology and Immunology , University of Saskatchewan , Saskatoon , Canada
| | - Ravendra Garg
- a VIDO-InterVac , University of Saskatchewan , Saskatoon , Canada
| | | |
Collapse
|
34
|
Omran GA. Hematological and immunological impairment following in-utero and postnatal exposure to aluminum sulfate in female offspring of albino rats. Immunopharmacol Immunotoxicol 2019; 41:40-47. [PMID: 30706732 DOI: 10.1080/08923973.2018.1533967] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Aim: Aluminum (Al) is a ubiquitous element extensively utilized in many products like food additives, pharmaceuticals, and vaccines, but its hematotoxic and immunotoxic effects are not entirely clarified. The present study explored the developmental hematotoxic and immunotoxic properties of aluminum sulfate (AS) in rats' offspring. Methods: Forty female offspring (10 rats each) were given three incremental AS doses plus a control group, from conception through lactation and after weaning until reached eight weeks old (near adults). Spleen relative weights along with total and differential blood counts were evaluated. Spectroscopic Al levels in spleen and brain were analyzed. Three immunoglobulins (IgG, IgM, and IgE) and two cytokines, interferon-γ and tumor necrosis factor-α, were measured through the ELISA technique. Results: The results revealed a significant relative increase in splenic weights mostly observed in the highest AS dose treated group. Reduction in the total leukocytic count was noticed in the three AS treated groups with relative lymphocytosis. Additionally, a significant decline in RBCs counts and hemoglobin concentrations were recorded. Tumor necrosis factor-α was significantly elevated in the three Al treated groups, while, interferon- γ showed a non-significant reduction compared to the control group. A significant increment in IgG and decline in IgE concentrations with no change in IgM level among groups were observed. Conclusion: Perinatal AS exposure caused mostly non-linear dose-dependent hematotoxicity and immunological impairment especially for the acquired immunity either cellular or humoral. Further studies can examine the immunotoxic effect of Al on male offspring during different stages of immune development.
Collapse
Affiliation(s)
- Ghada A Omran
- a Faculty of Medicine, Forensic Medicine and Clinical toxicology department , Assiut University , Assiut , Egypt
| |
Collapse
|
35
|
Chen W, Zuo H, Rolfe B, Schembri MA, Cobbold RN, Zhang B, Mahony TJ, Xu ZP. Clay nanoparticles co-deliver three antigens to promote potent immune responses against pathogenic Escherichia coli. J Control Release 2018; 292:196-209. [PMID: 30414464 DOI: 10.1016/j.jconrel.2018.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/02/2018] [Accepted: 11/04/2018] [Indexed: 01/02/2023]
|
36
|
Shardlow E, Mold M, Exley C. Unraveling the enigma: elucidating the relationship between the physicochemical properties of aluminium-based adjuvants and their immunological mechanisms of action. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2018; 14:80. [PMID: 30455719 PMCID: PMC6223008 DOI: 10.1186/s13223-018-0305-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/26/2018] [Indexed: 01/02/2023]
Abstract
Aluminium salts are by far the most commonly used adjuvants in vaccines. There are only two aluminium salts which are used in clinically-approved vaccines, Alhydrogel® and AdjuPhos®, while the novel aluminium adjuvant used in Gardasil® is a sulphated version of the latter. We have investigated the physicochemical properties of these two aluminium adjuvants and specifically in milieus approximating to both vaccine vehicles and the composition of injection sites. Additionally we have used a monocytic cell line to establish the relationship between their physicochemical properties and their internalisation and cytotoxicity. We emphasise that aluminium adjuvants used in clinically approved vaccines are chemically and biologically dissimilar with concomitantly potentially distinct roles in vaccine-related adverse events.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG UK
| | - Matthew Mold
- The Birchall Centre, Lennard Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG UK
| | - Christopher Exley
- The Birchall Centre, Lennard Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG UK
| |
Collapse
|
37
|
Shokouhi H, Farahmand B, Ghaemi A, Mazaheri V, Fotouhi F. Vaccination with three tandem repeats of M2 extracellular domain fused to Leismania major HSP70 protects mice against influenza A virus challenge. Virus Res 2018; 251:40-46. [DOI: 10.1016/j.virusres.2018.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 12/29/2022]
|
38
|
Shu F, Shi Y. Systematic Overview of Solid Particles and Their Host Responses. Front Immunol 2018; 9:1157. [PMID: 29892295 PMCID: PMC5985299 DOI: 10.3389/fimmu.2018.01157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/08/2018] [Indexed: 12/17/2022] Open
Abstract
Crystalline/particulate substances trigger a plethora of signaling events in host cells. The most prominent consequence is the inflammatory reactions that underlie crystal arthropathies, such as gout and pseudogout. However, their impact on our health was underestimated. Recent work on the role of cholesterol crystal in the development of atherosclerosis and the harm of environmental particulates has set up new frontiers in our defense against their detrimental effects. On the other hand, in the last 100 years, crystalline/particulate substances have been used with increasing frequencies in our daily lives as a part of new industrial manufacturing and engineering. Importantly, they have become a tool in modern medicine, used as vaccine adjuvants and drug delivery vehicles. Their biological effects are also being dissected in great detail, particularly with regard to their inflammatory signaling pathways. Solid structure interaction with host cells is far from being uniform, with outcomes dependent on cell types and chemical/physical properties of the particles involved. In this review, we offer a systematic and broad outlook of this landscape and a sage analysis of the complex nature of this topic.
Collapse
Affiliation(s)
- Fei Shu
- Department of Basic Medical Sciences, Institute for Immunology, Center for Life Sciences, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Peking University, Beijing, China
| | - Yan Shi
- Department of Basic Medical Sciences, Institute for Immunology, Center for Life Sciences, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
39
|
Terhune TD, Deth RC. Aluminum Adjuvant-Containing Vaccines in the Context of the Hygiene Hypothesis: A Risk Factor for Eosinophilia and Allergy in a Genetically Susceptible Subpopulation? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E901. [PMID: 29751492 PMCID: PMC5981940 DOI: 10.3390/ijerph15050901] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/19/2018] [Accepted: 04/29/2018] [Indexed: 12/17/2022]
Abstract
There are similarities between the immune response following immunization with aluminum adjuvants and the immune response elicited by some helminthic parasites, including stimulation of immunoglobulin E (IgE) and eosinophilia. Immunization with aluminum adjuvants, as with helminth infection, induces a Th2 type cell mediated immune response, including eosinophilia, but does not induce an environment conducive to the induction of regulatory mechanisms. Helminths play a role in what is known as the hygiene hypothesis, which proposes that decreased exposure to microbes during a critical time in early life has resulted in the increased prevalence and morbidity of asthma and atopic disorders over the past few decades, especially in Western countries. In addition, gut and lung microbiome composition and their interaction with the immune system plays an important role in a properly regulated immune system. Disturbances in microbiome composition are a risk factor for asthma and allergies. We propose that immunization with aluminum adjuvants in general is not favorable for induction of regulatory mechanisms and, in the context of the hygiene hypothesis and microbiome theory, can be viewed as an amplifying factor and significant contributing risk factor for allergic diseases, especially in a genetically susceptible subpopulation.
Collapse
Affiliation(s)
- Todd D Terhune
- College of Pharmacy, Department of Pharmaceutical Sciences, Nova Southeastern University, 1382 Terry Bldg, 3200 South University Drive, Fort Lauderdale, FL 33328, USA.
| | - Richard C Deth
- College of Pharmacy, Department of Pharmaceutical Sciences, Nova Southeastern University, 1382 Terry Bldg, 3200 South University Drive, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
40
|
Chen W, Zuo H, Li B, Duan C, Rolfe B, Zhang B, Mahony TJ, Xu ZP. Clay Nanoparticles Elicit Long-Term Immune Responses by Forming Biodegradable Depots for Sustained Antigen Stimulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1704465. [PMID: 29655306 DOI: 10.1002/smll.201704465] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/29/2018] [Indexed: 05/21/2023]
Abstract
Nanomaterials have been widely tested as new generation vaccine adjuvants, but few evoke efficient immunoreactions. Clay nanoparticles, for example, layered double hydroxide (LDH) and hectorite (HEC) nanoparticles, have shown their potent adjuvanticity in generating effective and durable immune responses. However, the mechanism by which clay nanoadjuvants stimulate the immune system is not well understood. Here, it is demonstrated that LDH and HEC-antigen complexes form loose agglomerates in culture medium/serum. They also form nodules with loose structures in tissue after subcutaneous injection, where they act as a depot for up to 35 d. More importantly, clay nanoparticles actively and continuously recruit immune cells into the depot for up to one month, and stimulate stronger immune responses than FDA-approved adjuvants, Alum and QuilA. Sustained antigen release is also observed in clay nanoparticle depots, with 50-60% antigen released after 35 d. In contrast, Alum-antigen complexes show minimal antigen release from the depot. Importantly, LDH and HEC are more effective than QuilA and Alum in promoting memory T-cell proliferation. These findings suggest that both clay nanoadjuvants can serve as active vaccine platforms for sustained and potent immune responses.
Collapse
Affiliation(s)
- Weiyu Chen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Huali Zuo
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Bei Li
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Chengcheng Duan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Barbara Rolfe
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Bing Zhang
- Vaccine Delivery, Animal Science, Agri-Science Queensland, Department of Agriculture & Fisheries, Dutton Park, QLD, 4102, Australia
| | - Timothy J Mahony
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| |
Collapse
|
41
|
Katikaneni DS, Jin L. B cell MHC class II signaling: A story of life and death. Hum Immunol 2018; 80:37-43. [PMID: 29715484 DOI: 10.1016/j.humimm.2018.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/08/2018] [Accepted: 04/25/2018] [Indexed: 01/17/2023]
Abstract
MHC class II regulates B cell activation, proliferation, and differentiation during cognate B cell-T cell interaction. This is, in part, due to the MHC class II signaling in B cells. Activation of MHC Class II in human B cells or "primed" murine B cells leads to tyrosine phosphorylation, calcium mobilization, AKT, ERK, JNK activation. In addition, crosslinking MHC class II with monoclonal Abs kill malignant human B cells. Several humanized anti-HLA-DR/MHC class II monoclonal Abs entered clinical trials for lymphoma/leukemia and MHC class II-expressing melanomas. Mechanistically, MHC class II is associated with a wealth of transmembrane proteins including the B cell-specific signaling proteins CD79a/b, CD19 and a group of four-transmembrane proteins including tetraspanins and the apoptotic protein MPYS/STING. Furthermore, MHC class II signals are compartmentalized in the tetraspanin-enriched microdomains. In this review, we discuss our current understanding of MHC class II signaling in B cells focusing on its physiological significance and the therapeutic potential.
Collapse
Affiliation(s)
- Divya Sai Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, United States
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
42
|
Shen M, Wang J, Yu W, Zhang C, Liu M, Wang K, Yang L, Wei F, Wang SE, Sun Q, Ren X. A novel MDSC-induced PD-1 -PD-L1 + B-cell subset in breast tumor microenvironment possesses immuno-suppressive properties. Oncoimmunology 2018; 7:e1413520. [PMID: 29632731 DOI: 10.1080/2162402x.2017.1413520] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/30/2017] [Accepted: 11/30/2017] [Indexed: 10/18/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells that suppress T-cell activity in a tumor microenvironment. However, the suppressive function of MDSCs on B cells and its underlying mechanism remain unclear. Here, we show that in 4T1 breast cancer mice, a significantly increased number of MDSCs, in parallel with splenic B cells, are accumulated when compared to normal mice. In the presence of MDSCs, the surface molecules of B cells are remolded, with checkpoint-related molecules such as PD-1 and PD-L1 changing prominently. MDSCs also emerge as vital regulators in B-cell immune functions such as proliferation, apoptosis and the abilities to secrete antibodies and cytokines. Our study further identifies that MDSCs can transform normal B cells to a subtype of immuno- regulatory B cells (Bregs) which inhibit T-cell response. Furthermore, we identified a novel kind of Bregs with a specific phenotype PD-1-PD-L1+CD19+, which exert the greatest suppressive effects on T cells in comparison with the previously reported Bregs characterized as CD1d+CD5+CD19+, CD5+CD19+ and Interleukin (IL)-10-secreting B cells. Our results highlight that MDSCs regulate B-cell response and may serve as a therapeutic approach in anti-tumor treatment. Investigation of this new Breg subtype extends our understanding of regulation of T-cell response and sheds new light on anti-tumor immunity and immune therapy.
Collapse
Affiliation(s)
- Meng Shen
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Chen Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Min Liu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Kaiyuan Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Shizhen Emily Wang
- Department of Pathology, University of California, San Diego, California, USA
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| |
Collapse
|
43
|
Abstract
Eosinophils are a prominent cell type in particular host responses such as the response to helminth infection and allergic disease. Their effector functions have been attributed to their capacity to release cationic proteins stored in cytoplasmic granules by degranulation. However, eosinophils are now being recognized for more varied functions in previously underappreciated diverse tissue sites, based on the ability of eosinophils to release cytokines (often preformed) that mediate a broad range of activities into the local environment. In this Review, we consider evolving insights into the tissue distribution of eosinophils and their functional immunobiology, which enable eosinophils to secrete in a selective manner cytokines and other mediators that have diverse, 'non-effector' functions in health and disease.
Collapse
Affiliation(s)
- Peter F Weller
- Division of Allergy and Inflammation, Harvard Medical School, Beth Israel Deaconess Medical Center, CLS 943, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Lisa A Spencer
- Division of Allergy and Inflammation, Harvard Medical School, Beth Israel Deaconess Medical Center, CLS 943, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| |
Collapse
|
44
|
Unperturbed Immune Function despite Mutation of C-Terminal Tyrosines in Syk Previously Implicated in Signaling and Activity Regulation. Mol Cell Biol 2017; 37:MCB.00216-17. [PMID: 28760774 DOI: 10.1128/mcb.00216-17] [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: 04/28/2017] [Accepted: 07/26/2017] [Indexed: 11/20/2022] Open
Abstract
The nonreceptor tyrosine kinase Syk, a central regulator of immune cell differentiation and activation, is a promising drug target for treatment of leukemia and allergic and inflammatory diseases. The clinical failure of Syk inhibitors underscores the importance of understanding the regulation of Syk function and activity. A series of previous studies emphasized the importance of three C-terminal tyrosines in Syk for kinase activity regulation, as docking sites for downstream effector molecules, and for Ca2+ mobilization. Here, we investigated the roles of these C-terminal tyrosines in the mouse. Surprisingly, expression of a triple tyrosine-to-phenylalanine human Syk mutant, SYK(Y3F), was not associated with discernible signaling defects either in reconstituted DT40 cells or in B or mast cells from mice expressing SYK(Y3F) instead of wild-type Syk. Remarkably, lymphocyte differentiation, calcium mobilization, and 2,4,6-trinitrophenyl (TNP)-specific immune responses were unperturbed in SYK(Y3F) mice. These results emphasize the capacity of immune cells to compensate for specific molecular defects, likely using redundant intermolecular interactions, and highlight the importance of in vivo analyses for understanding cellular signaling mechanisms.
Collapse
|
45
|
Efficient induction of comprehensive immune responses to control pathogenic E. coli by clay nano-adjuvant with the moderate size and surface charge. Sci Rep 2017; 7:13367. [PMID: 29042573 PMCID: PMC5645426 DOI: 10.1038/s41598-017-13570-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/26/2017] [Indexed: 02/06/2023] Open
Abstract
In recent decades, diseases caused by pathogenic Escherichia coli (E. coli), enterohaemorrhagic E. coli (EHEC) O26 have been increasingly reported worldwide, which are as severe as those caused by EHEC strain O157:H7 and require effective intervention strategies. Herein, we report the application of clay nanoparticles, i.e. hectorites as effective nano-adjuvants for vaccination against EHEC O26 colonization. We show that medium size HEC (hectorite, around 73~77 nm diameter) is able to induce efficient humoral and cellular immune responses against EHEC antigen - intimin β (IB), which are significantly higher than those triggered by commercially used adjuvants - QuilA and Alum. We also demonstrate that mice immunized with IB adjuvanted with HEC nanoparticles elicit sufficient secretion of mucosal IgA, capable of providing effective protection against EHEC O26 binding to ruminant and human cells. In addition, we demonstrate for the first time that hectorites are able to initiate maturation of RAW 264.7 macrophages, inducing expression of co-stimulatory cytokines at a low nanoparticle concentration (10 μg/mL). Together these data strongly suggest that hectorite with optimized size is a highly efficient vaccine nano-adjuvant.
Collapse
|
46
|
Mion F, Vetrano S, Tonon S, Valeri V, Piontini A, Burocchi A, Petti L, Frossi B, Gulino A, Tripodo C, Colombo MP, Pucillo CE. Reciprocal influence of B cells and tumor macro and microenvironments in the ApcMin/+ model of colorectal cancer. Oncoimmunology 2017; 6:e1336593. [PMID: 28919998 DOI: 10.1080/2162402x.2017.1336593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 12/22/2022] Open
Abstract
One of the most fascinating aspects of the immune system is its dynamism, meant as the ability to change and readapt according to the organism needs. Following an insult, we assist to the spontaneous organization of different immune cells which cooperate, locally and at distance, to build up an appropriate response. Throughout tumor progression, adaptations within the systemic tumor environment, or macroenvironment, result in the promotion of tumor growth, tumor invasion and metastasis to distal organs, but also to dramatic changes in the activity and composition of the immune system. In this work, we show the changes of the B-cell arm of the immune system following tumor progression in the ApcMin/+ model of colorectal cancer. Tumor macroenvironment leads to an increased proportion of total and IL-10-competent B cells in draining LNs while activates a differentiation route that leads to the expansion of IgA+ lymphocytes in the spleen and peritoneum. Importantly, serum IgA levels were significantly higher in ApcMin/+ than Wt mice. The peculiar involvement of IgA response in the adenomatous transformation had correlates in the gut-mucosal compartment where IgA-positive elements increased from normal mucosa to areas of low grade dysplasia while decreasing upon overt carcinomatous transformation. Altogether, our findings provide a snapshot of the tumor education of B lymphocytes in the ApcMin/+ model of colorectal cancer. Understanding how tumor macroenvironment affects the differentiation, function and distribution of B lymphocytes is pivotal to the generation of specific therapies, targeted to switching B cells to an anti-, rather than pro-, tumoral phenotype.
Collapse
Affiliation(s)
- Francesca Mion
- Department of Medicine, University of Udine, Udine, Italy
| | - Stefania Vetrano
- Inflammatory Bowel Disease Center, Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Silvia Tonon
- Department of Medicine, University of Udine, Udine, Italy
| | - Viviana Valeri
- Department of Medicine, University of Udine, Udine, Italy
| | - Andrea Piontini
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Alessia Burocchi
- Department of Experimental Oncology and Molecular Medicine, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Luciana Petti
- Department of Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Barbara Frossi
- Department of Medicine, University of Udine, Udine, Italy
| | - Alessandro Gulino
- Department of Health Science, Tumor Immunology Unit, Human Pathology Section, Palermo University School of Medicine, Palermo, Italy
| | - Claudio Tripodo
- Department of Health Science, Tumor Immunology Unit, Human Pathology Section, Palermo University School of Medicine, Palermo, Italy
| | - Mario P Colombo
- Department of Experimental Oncology and Molecular Medicine, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | | |
Collapse
|
47
|
Malm M, Heinimäki S, Vesikari T, Blazevic V. Rotavirus capsid VP6 tubular and spherical nanostructures act as local adjuvants when co-delivered with norovirus VLPs. Clin Exp Immunol 2017; 189:331-341. [PMID: 28407442 PMCID: PMC5543502 DOI: 10.1111/cei.12977] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2017] [Indexed: 01/19/2023] Open
Abstract
A subunit protein vaccine candidate based on norovirus (NoV) virus‐like particles (VLPs) and rotavirus (RV) VP6 protein against acute childhood gastroenteritis has been proposed recently. RV VP6 forms different oligomeric nanostructures, including tubes and spheres when expressed in vitro, which are highly immunogenic in different animal models. We have shown recently that recombinant VP6 nanotubes have an adjuvant effect on immunogenicity of NoV VLPs in mice. In this study, we investigated if the adjuvant effect is dependent upon a VP6 dose or different VP6 structural assemblies. In addition, local and systemic adjuvant effects as well as requirements for antigen co‐delivery and co‐localization were studied. The magnitude and functionality of NoV GII.4‐specific antibodies and T cell responses were tested in mice immunized with GII.4 VLPs alone or different combinations of VLPs and VP6. A VP6 dose‐dependent adjuvant effect on GII.4‐specific antibody responses was observed. The adjuvant effect was found to be strictly dependent upon co‐administration of NoV GII.4 VLPs and VP6 at the same anatomic site and at the same time. However, the adjuvant effect was not dependent on the types of oligomers used, as both nanotubes and nanospheres exerted adjuvant effect on GII.4‐specific antibody generation and, for the first time, T cell immunity. These findings elucidate the mechanisms of VP6 adjuvant effect in vivo and support its use as an adjuvant in a combination NoV and RV vaccine.
Collapse
Affiliation(s)
- M Malm
- Vaccine Research Center, University of Tampere, Tampere, Finland
| | - S Heinimäki
- Vaccine Research Center, University of Tampere, Tampere, Finland
| | - T Vesikari
- Vaccine Research Center, University of Tampere, Tampere, Finland
| | - V Blazevic
- Vaccine Research Center, University of Tampere, Tampere, Finland
| |
Collapse
|
48
|
Gnopo YM, Watkins HC, Stevenson TC, DeLisa MP, Putnam D. Designer outer membrane vesicles as immunomodulatory systems - Reprogramming bacteria for vaccine delivery. Adv Drug Deliv Rev 2017; 114:132-142. [PMID: 28501509 DOI: 10.1016/j.addr.2017.05.003] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 12/19/2022]
Abstract
Vaccines often require adjuvants to be effective. Traditional adjuvants, like alum, activate the immune response but in an uncontrolled way. Newer adjuvants help to direct the immune response in a more coordinated fashion. Here, we review the opportunity to use the outer membrane vesicles (OMVs) of bacteria as a way to modulate the immune response toward making more effective vaccines. This review outlines the different types of OMVs that have been investigated for vaccine delivery and how they are produced. Because OMVs are derived from bacteria, they have compositions that may not be compatible with parenteral delivery in humans; therefore, we also review the strategies brought to bear to detoxify OMVs while maintaining an adjuvant profile. OMV-based vaccines can be derived from the pathogens themselves, or can be used as surrogate constructs to mimic a pathogen through the heterologous expression of specific antigens in a desired host source strain, and approaches to doing so are reviewed. Additionally, the emerging area of engineered pathogen-specific carbohydrate sequences, or glycosylated OMVs is reviewed and contrasted with protein antigen delivery. Existing OMV-based vaccines as well as their routes of administration round out the text. Overall, this is an exciting time in the OMV field as it matures and leads to more effective and targeted ways to induce desired pathogen-specific immune responses.
Collapse
|
49
|
Shardlow E, Mold M, Exley C. From Stock Bottle to Vaccine: Elucidating the Particle Size Distributions of Aluminum Adjuvants Using Dynamic Light Scattering. Front Chem 2017; 4:48. [PMID: 28119911 PMCID: PMC5220009 DOI: 10.3389/fchem.2016.00048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/19/2016] [Indexed: 12/30/2022] Open
Abstract
The physicochemical properties of aluminum salts are key determinants of their resultant adjuvanticity in vivo when administered as part of a vaccine. While there are links between particle size and the efficacy of the immune response, the limited literature directly characterizing the PSD of aluminum adjuvants has stymied the elucidation of such a relationship for these materials. Hence, this comparative study was undertaken to monitor the PSD of aluminum adjuvants throughout the process of vaccine formulation using DLS. A significant proportion of the stock suspensions was highly agglomerated (>9 μm) and Alhydrogel® exhibited the smallest median size (2677 ± 120 nm) in comparison to Adju-Phos® or Imject alum® (7152 ± 308 and 7294 ± 146 nm respectively) despite its large polydispersity index (PDI). Dilution of these materials induced some degree of disaggregation within all samples with Adju-Phos® being the most significantly affected. The presence of BSA caused the median size of Alhydrogel® to increase but these trends were not evident when model vaccines were formulated with either Adju-Phos® or Imject alum®. Nevertheless, Alhydrogel® and Adju-Phos® exhibited comparable median sizes in the presence of this protein (4194 ± 466 and 4850 ± 501 nm respectively) with Imject alum® being considerably smaller (2155 ± 485 nm). These results suggest that the PSD of aluminum adjuvants is greatly influenced by dilution and the degree of protein adsorption experienced within the vaccine itself. The size of the resultant antigen-adjuvant complex may be important for its immunological recognition and subsequent clearance from the injection site.
Collapse
Affiliation(s)
- Emma Shardlow
- Lennard-Jones Laboratories, The Birchall Centre, Keele University Staffordshire, UK
| | - Matthew Mold
- Lennard-Jones Laboratories, The Birchall Centre, Keele University Staffordshire, UK
| | - Christopher Exley
- Lennard-Jones Laboratories, The Birchall Centre, Keele University Staffordshire, UK
| |
Collapse
|
50
|
Artemisinin analogue SM934 attenuate collagen-induced arthritis by suppressing T follicular helper cells and T helper 17 cells. Sci Rep 2016; 6:38115. [PMID: 27897259 PMCID: PMC5126690 DOI: 10.1038/srep38115] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/04/2016] [Indexed: 12/20/2022] Open
Abstract
SM934 is an artemisinin analogue with immunosuppressive properties and potent therapeutic activity against lupus-like diseases in autoimmune mice. In this report, the therapeutic efficacy and underlying mechanisms of SM934 on rheumatoid arthritis (RA) was investigated using collagen-induced arthritis (CIA) in DBA/1J mice. We demonstrated that SM934 treatment alleviate the severity of arthritis in CIA mice with established manifestations. The therapeutic benefits were associated with ameliorated joint swelling and reduced extent of bone erosion and destruction. Further, administration of SM934 diminished the development of T follicular helper (Tfh) cells and Th17 cells and suppressed the production of pathogenic antibodies, without altering the proportion of germinal center B cells. Ex vivo, SM934 treatment inhibited the bovine type II collagen (CII) induced proliferation and inflammatory cytokines secretion of CII -reactive T cells. In vitro, SM934 impeded the polarization of naïve CD4+ T cells into Tfh cells and the expression of its transcript factor Bcl-6. Moreover, SM934 decreased the IL-21-producing CD4+ T cells and dampened the IL-21 downstream signaling through STAT3. These finding offered the convincing evidence that artemisinin derivative might attenuate RA by simultaneously interfering with the generation of Tfh cells and Th17 cells as well as the subsequent antibody-mediated immune responses.
Collapse
|