1
|
Pellegrina D, Wilson HL, Mutwiri GK, Helmy M. Transcriptional Systems Vaccinology Approaches for Vaccine Adjuvant Profiling. Vaccines (Basel) 2025; 13:33. [PMID: 39852812 PMCID: PMC11768747 DOI: 10.3390/vaccines13010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Adjuvants are a diverse group of substances that can be added to vaccines to enhance antigen-specific immune responses and improve vaccine efficacy. The first adjuvants, discovered almost a century ago, were soluble crystals of aluminium salts. Over the following decades, oil emulsions, vesicles, oligodeoxynucleotides, viral capsids, and other complex organic structures have been shown to have adjuvant potential. However, the detailed mechanisms of how adjuvants enhance immune responses remain poorly understood and may be a barrier that reduces the rational selection of vaccine components. Previous studies on mechanisms of action of adjuvants have focused on how they activate innate immune responses, including the regulation of cell recruitment and activation, cytokine/chemokine production, and the regulation of some "immune" genes. This approach provides a narrow perspective on the complex events involved in how adjuvants modulate antigen-specific immune responses. A comprehensive and efficient way to investigate the molecular mechanism of action for adjuvants is to utilize systems biology approaches such as transcriptomics in so-called "systems vaccinology" analysis. While other molecular biology methods can verify if one or few genes are differentially regulated in response to vaccination, systems vaccinology provides a more comprehensive picture by simultaneously identifying the hundreds or thousands of genes that interact with complex networks in response to a vaccine. Transcriptomics tools such as RNA sequencing (RNA-Seq) allow us to simultaneously quantify the expression of practically all expressed genes, making it possible to make inferences that are only possible when considering the system as a whole. Here, we review some of the challenges in adjuvant studies, such as predicting adjuvant activity and toxicity when administered alone or in combination with antigens, or classifying adjuvants in groups with similar properties, while underscoring the significance of transcriptomics in systems vaccinology approaches to propel vaccine development forward.
Collapse
Affiliation(s)
- Diogo Pellegrina
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
| | - Heather L. Wilson
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
- Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - George K. Mutwiri
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
- Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Mohamed Helmy
- Vaccine and Infectious Diseases Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada; (D.P.); (H.L.W.); (G.K.M.)
- Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Computer Science, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Computer Science, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
- Department of Computer Science, Idaho State University, Pocatello, ID 83209, USA
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
| |
Collapse
|
2
|
González-Domínguez I, Puente-Massaguer E, Abdeljawad A, Lai TY, Liu Y, Loganathan M, Francis B, Lemus N, Dolange V, Boza M, Slamanig S, Martínez-Guevara JL, Krammer F, Palese P, Sun W. Preclinical evaluation of a universal inactivated influenza B vaccine based on the mosaic hemagglutinin-approach. NPJ Vaccines 2024; 9:222. [PMID: 39551795 PMCID: PMC11570629 DOI: 10.1038/s41541-024-01014-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
We have developed a new universal influenza B vaccination strategy based on inactivated influenza B viruses displaying mosaic hemagglutinins (mHAs). Recombinant mHA viruses were constructed by replacing the four major antigenic sites of influenza B virus HAs, with those from exotic avian influenza A virus HAs. Sequential vaccination of naïve mice with mHA-based vaccines elicited higher immune responses towards the immuno-subdominant conserved epitopes of the HA than vaccination with wildtype viruses. Among the different preparations tested, mHA split vaccines were less immunogenic than their whole inactivated virus counterparts. This lower immunogenicity was overcome by the combination with adjuvants. mHA split vaccines adjuvanted with a Toll-like receptor-9 agonist (CpG 1018) increased Th1 immunity and in vivo cross-protection, whereas adjuvanting with an MF59-like oil-in-water nano-emulsion (AddaVax) enhanced and broadened humoral immune responses and antibody-mediated cross-protection. The mHA vaccines with or without adjuvant were subsequently evaluated in mice that were previously immunized to closely mimic human pre-existing immunity to influenza B viruses and the contribution of innate and cellular immunity was evaluated in this model. We believe these preclinical studies using the mHA strategy represent a major step toward the evaluation of a universal influenza B virus vaccine in clinical trials.
Collapse
Affiliation(s)
| | - Eduard Puente-Massaguer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Adam Abdeljawad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tsoi Ying Lai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yonghong Liu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Benjamin Francis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicholas Lemus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Victoria Dolange
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marta Boza
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stefan Slamanig
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
3
|
Blass E, Colarusso A, Aid M, Larocca RA, Reeves RK, Barouch DH. Early spatiotemporal evolution of the immune response elicited by adenovirus serotype 26 vector vaccination in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.618988. [PMID: 39464013 PMCID: PMC11507988 DOI: 10.1101/2024.10.18.618988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
As the first responder to immunological challenges, the innate immune system shapes and regulates the ensuing adaptive immune response. Many clinical studies evaluating the role of innate immunity in initiating vaccine-elicited adaptive immune responses have largely been confined to blood due to inherent difficulty in acquiring tissue samples. However, the absence of vaccine-site and draining lymph node information limits understanding of early events induced by vaccination that could potentially shape vaccine-elicited immunity. We therefore utilized a mouse model to investigate the spatiotemporal evolution of the immune response within the first 24 hours following intramuscular adenovirus serotype 26 (Ad26) vector vaccination in tissues. We show that the Ad26 vaccine-elicited innate immune response commences by one hour and rapidly evolves in tissues and blood within the first 24 hours as reflected by the detection of cytokines, chemokines, cellular responses, and transcriptomic pathways. Furthermore, serum levels of IL-6, MIG, MIP-1α, and MIP-1β at 6 hours post-vaccination correlated with the frequency of vaccine-elicited memory CD8+ T cell responses evaluated at 60 days post-vaccination in blood and tissues. Taken together, our data suggests that the immune response to Ad26 vector vaccination commences quickly in tissues by one hour and that events by as early as 6 hours post-vaccination can shape vaccine-elicited CD8+ T cell responses at later memory time points.
Collapse
Affiliation(s)
- Eryn Blass
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Alessandro Colarusso
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Rafael A. Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - R. Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
4
|
Liu A, Liu G, Wang X, Yan D, Zhang J, Wei L. Comprehensive analysis of single-cell RNA and bulk RNA sequencing based on M2 tumor-associated macrophage and angiogenesis-related genes to assess prognosis and therapeutic response in lung adenocarcinoma. Heliyon 2024; 10:e34784. [PMID: 39148979 PMCID: PMC11325380 DOI: 10.1016/j.heliyon.2024.e34784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
M2 tumor-associated macrophage (M2 TAM), a crucial component of the tumor microenvironment, has a significant impact on tumor invasion and metastasis in the form of angiogenesis for lung adenocarcinoma (LUAD). In this study, both single-cell RNA and bulk RNA sequencing data were analyzed to identify 12 M2 TAM and angiogenesis-related genes (OLR1, CTSL, HLA-DPB1, NUPR1, ALOX5, DOCK4, CSF2RB, PTPN6, TNFSF12, HNRNPA2B1, NCL, and BIRC2). These genes were used to construct a prognostic signature, which was subsequently validated using an external cohort. Moreover, the immune profile analysis indicated that the low-risk group exhibited a distinct immune cell infiltration and relatively active status. Importantly, the prognostic signature was closely associated with PD-1, CTLA4, tumor mutation burden, and anti-cancer drug sensitivity. In summary, this study proposes a new prognostic signature for patients with LUAD based on M2 TAM and angiogenesis-related genes. The signature forecasts the prognosis of LUAD by an independent manner, reveals the potential molecular mechanisms involved in tumor immune-related functions, and offers appropriate clinical strategies for the treatment of patients with LUAD.
Collapse
Affiliation(s)
- Anbang Liu
- Department of Thoracic Surgery, Qingdao Municipal Hospital, 266000, Qingdao, Shandong, China
| | - Gengqiu Liu
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming Dist., Shenzhen, 518107, China
| | - Xiaohuai Wang
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming Dist., Shenzhen, 518107, China
| | - Dongqing Yan
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming Dist., Shenzhen, 518107, China
| | - Junhang Zhang
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming Dist., Shenzhen, 518107, China
| | - Li Wei
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming Dist., Shenzhen, 518107, China
| |
Collapse
|
5
|
Davies ML, Biryukov SS, Rill NO, Klimko CP, Hunter M, Dankmeyer JL, Miller JA, Shoe JL, Mlynek KD, Talyansky Y, Toothman RG, Qiu J, Bozue JA, Cote CK. Sex differences in immune protection in mice conferred by heterologous vaccines for pneumonic plague. Front Immunol 2024; 15:1397579. [PMID: 38835755 PMCID: PMC11148226 DOI: 10.3389/fimmu.2024.1397579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/25/2024] [Indexed: 06/06/2024] Open
Abstract
Background Yersinia pestis is the etiological agent of plague, which can manifest as bubonic, septicemic, and/or pneumonic disease. Plague is a severe and rapidly progressing illness that can only be successfully treated with antibiotics initiated early after infection. There are no FDA-approved vaccines for plague, and some vaccine candidates may be less effective against pneumonic plague than bubonic plague. Y. pestis is not known to impact males and females differently in mechanisms of pathogenesis or severity of infection. However, one previous study reported sex-biased vaccine effectiveness after intranasal Y. pestis challenge. As part of developing a safe and effective vaccine, it is essential that potential sex differences are characterized. Methods In this study we evaluated novel vaccines in male and female BALB/c mice using a heterologous prime-boost approach and monitored survival, bacterial load in organs, and immunological correlates. Our vaccine strategy consisted of two subcutaneous immunizations, followed by challenge with aerosolized virulent nonencapsulated Y. pestis. Mice were immunized with a combination of live Y. pestis pgm- pPst-Δcaf1, live Y. pestis pgm- pPst-Δcaf1/ΔyopD, or recombinant F1-V (rF1-V) combined with adjuvants. Results The most effective vaccine regimen was initial priming with rF1-V, followed by boost with either of the live attenuated strains. However, this and other strategies were more protective in female mice. Males had higher bacterial burden and differing patterns of cytokine expression and serum antibody titers. Male mice did not demonstrate synergy between vaccination and antibiotic treatment as repeatedly observed in female mice. Conclusions This study provides new knowledge about heterologous vaccine strategies, sex differences in plague-vaccine efficacy, and the immunological factors that differ between male and female mice.
Collapse
Affiliation(s)
- Michael L Davies
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Sergei S Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Nathaniel O Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jennifer L Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jeremy A Miller
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Kevin D Mlynek
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Yuli Talyansky
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Ronald G Toothman
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Ju Qiu
- Regulated Research Administration: Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Joel A Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| |
Collapse
|
6
|
Ribette T, Charretier Y, Laurent S, Syntin P, Chautard E, Meniche X, Darnaud M, Bequet F, Beloeil L, Piras-Douce F, Abi-Ghanem J. Development of Mass Spectrometry Imaging on skeletal muscle to characterize the local pro-inflammatory and pro-resolution lipid responses in a vaccination context. J Proteomics 2024; 296:105105. [PMID: 38325731 DOI: 10.1016/j.jprot.2024.105105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/09/2024]
Abstract
Vaccine reactogenicity is well documented at the clinical level but the mechanism involved at the local or systemic level are still poorly understood. Muscular tissue where most vaccines are administered is the first place of interaction between the vaccine formulation and the host's immune cells. So far, this site of vaccine administration is not well documented from a mechanistic standpoint. The study of early molecular events at the injection site is crucial to understand the local response to vaccines. In this paper, we report a standardized workflow, from the injection of vaccine formulations in rabbit muscle, to the analysis by desorption electrospray ionization and histology staining to understand the role of lipids involved in the inflammation and its resolution on striated muscular tissue. The analysis of lipid mediators was optimized at the site of needle insertion to allow the spatial comparison of cellular infiltrates at the injection site. We showed that lipids were distributed across the spatial tissue morphology in a time-dependent manner. The MS imaging applied to vaccinology could pave the way to a better understanding of vaccine reactogenicity and mechanism of action.
Collapse
|
7
|
Gonzalez Dias Carvalho PC, Dominguez Crespo Hirata T, Mano Alves LY, Moscardini IF, do Nascimento APB, Costa-Martins AG, Sorgi S, Harandi AM, Ferreira DM, Vianello E, Haks MC, Ottenhoff THM, Santoro F, Martinez-Murillo P, Huttner A, Siegrist CA, Medaglini D, Nakaya HI. Baseline gene signatures of reactogenicity to Ebola vaccination: a machine learning approach across multiple cohorts. Front Immunol 2023; 14:1259197. [PMID: 38022684 PMCID: PMC10663260 DOI: 10.3389/fimmu.2023.1259197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The rVSVDG-ZEBOV-GP (Ervebo®) vaccine is both immunogenic and protective against Ebola. However, the vaccine can cause a broad range of transient adverse reactions, from headache to arthritis. Identifying baseline reactogenicity signatures can advance personalized vaccinology and increase our understanding of the molecular factors associated with such adverse events. Methods In this study, we developed a machine learning approach to integrate prevaccination gene expression data with adverse events that occurred within 14 days post-vaccination. Results and Discussion We analyzed the expression of 144 genes across 343 blood samples collected from participants of 4 phase I clinical trial cohorts: Switzerland, USA, Gabon, and Kenya. Our machine learning approach revealed 22 key genes associated with adverse events such as local reactions, fatigue, headache, myalgia, fever, chills, arthralgia, nausea, and arthritis, providing insights into potential biological mechanisms linked to vaccine reactogenicity.
Collapse
Affiliation(s)
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Leandro Yukio Mano Alves
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - André G. Costa-Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Artificial Intelligence and Analytics Department, Institute for Technological Research, São Paulo, Brazil
| | - Sara Sorgi
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ali M. Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Daniela M. Ferreira
- Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Angela Huttner
- Centre for Vaccinology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Infectious Diseases Service, Geneva University Hospitals, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Centre for Vaccinology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Donata Medaglini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Helder I. Nakaya
- Scientific Platform Pasteur-University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
8
|
Doss-Gollin S, Thomas S, Brook B, Abedi K, Lebas C, Auderset F, Lugo-Rodriguez Y, Sanchez-Schmitz G, Dowling DJ, Levy O, van Haren SD. Human in vitro modeling of adjuvant formulations demonstrates enhancement of immune responses to SARS-CoV-2 antigen. NPJ Vaccines 2023; 8:163. [PMID: 37884538 PMCID: PMC10603059 DOI: 10.1038/s41541-023-00759-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Adjuvants can enhance vaccine immunogenicity, but their mechanism of action is often incompletely understood, hampering rapid applicability for pandemic vaccines. Herein, we characterized the cellular and molecular activity of adjuvant formulations available for pre-clinical evaluation, including several developed for global open access. We applied four complementary human in vitro platforms to assess individual and combined adjuvants in unformulated, oil-in-water, and liposomal delivery platforms. Liposomal co-formulation of MPLA and QS-21 was most potent in promoting dendritic cell maturation, selective production of Th1-polarizing cytokines, and activation of SARS-CoV-2 Spike-specific CD4+ and CD8+ T cells in a co-culture assay. Select formulations also significantly enhanced Spike antigen-specific humoral immunity in vivo. This study confirms the utility of the cumulative use of human in vitro tools to predict adjuvanticity potential. Thus, human in vitro modeling may advance public health by accelerating the development of affordable and scalable adjuvants for vaccines tailored to vulnerable populations.
Collapse
Affiliation(s)
- Simon Doss-Gollin
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Sanya Thomas
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Byron Brook
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kimia Abedi
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Célia Lebas
- Vaccine Formulation Institute, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Floriane Auderset
- Vaccine Formulation Institute, 1228 Plan-les-Ouates, Geneva, Switzerland
| | | | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
9
|
Tregoning JS, Stirling DC, Wang Z, Flight KE, Brown JC, Blakney AK, McKay PF, Cunliffe RF, Murugaiah V, Fox CB, Beattie M, Tam YK, Johansson C, Shattock RJ. Formulation, inflammation, and RNA sensing impact the immunogenicity of self-amplifying RNA vaccines. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:29-42. [PMID: 36589712 PMCID: PMC9794906 DOI: 10.1016/j.omtn.2022.11.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
To be effective, RNA vaccines require both in situ translation and the induction of an immune response to recruit cells to the site of immunization. These factors can pull in opposite directions with the inflammation reducing expression of the vaccine antigen. We investigated how formulation affects the acute systemic cytokine response to a self-amplifying RNA (saRNA) vaccine. We compared a cationic polymer (pABOL), a lipid emulsion (nanostructured lipid carrier, NLC), and three lipid nanoparticles (LNP). After immunization, we measured serum cytokines and compared the response to induced antibodies against influenza virus. Formulations that induced a greater cytokine response induced a greater antibody response, with a significant correlation between IP-10, MCP-1, KC, and antigen-specific antibody titers. We then investigated how innate immune sensing and signaling impacted the adaptive immune response to vaccination with LNP-formulated saRNA. Mice that lacked MAVS and are unable to signal through RIG-I-like receptors had an altered cytokine response to saRNA vaccination and had significantly greater antibody responses than wild-type mice. This indicates that the inflammation induced by formulated saRNA vaccines is not solely deleterious in the induction of antibody responses and that targeting specific aspects of RNA vaccine sensing might improve the quality of the response.
Collapse
Affiliation(s)
- John S. Tregoning
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - David C. Stirling
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Katie E. Flight
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Jonathan C. Brown
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Anna K. Blakney
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Paul F. McKay
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Robert F. Cunliffe
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Valarmathy Murugaiah
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| | - Christopher B. Fox
- IDRI, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Mitchell Beattie
- Acuitas Therapeutics, 6190 Agronomy Road, Ste 405, Vancouver, BC, Canada
| | - Ying K. Tam
- Acuitas Therapeutics, 6190 Agronomy Road, Ste 405, Vancouver, BC, Canada
| | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, St. Mary’s Campus, London, UK
| | - Robin J. Shattock
- Department of Infectious Disease, Imperial College London, St. Mary’s Campus, London, UK
| |
Collapse
|
10
|
Identification and Application of a Novel Immune-Related lncRNA Signature on the Prognosis and Immunotherapy for Lung Adenocarcinoma. Diagnostics (Basel) 2022; 12:diagnostics12112891. [PMID: 36428951 PMCID: PMC9689875 DOI: 10.3390/diagnostics12112891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Long non-coding RNA (lncRNA) participates in the immune regulation of lung cancer. However, limited studies showed the potential roles of immune-related lncRNAs (IRLs) in predicting survival and immunotherapy response of lung adenocarcinoma (LUAD). Methods: Based on The Cancer Genome Atlas (TCGA) and ImmLnc databases, IRLs were identified through weighted gene coexpression network analysis (WGCNA), Cox regression, and Lasso regression analyses. The predictive ability was validated by Kaplan−Meier (KM) and receiver operating characteristic (ROC) curves in the internal dataset, external dataset, and clinical study. The immunophenoscore (IPS)-PD1/PD-L1 blocker and IPS-CTLA4 blocker data of LUAD were obtained in TCIA to predict the response to immune checkpoint inhibitors (ICIs). The expression levels of immune checkpoint molecules and markers for hyperprogressive disease were analyzed. Results: A six-IRL signature was identified, and patients were stratified into high- and low-risk groups. The low-risk had improved survival outcome (p = 0.006 in the training dataset, p = 0.010 in the testing dataset, p < 0.001 in the entire dataset), a stronger response to ICI (p < 0.001 in response to anti-PD-1/PD-L1, p < 0.001 in response to anti-CTLA4), and higher expression levels of immune checkpoint molecules (p < 0.001 in PD-1, p < 0.001 in PD-L1, p < 0.001 in CTLA4) but expressed more biomarkers of hyperprogression in immunotherapy (p = 0.002 in MDM2, p < 0.001 in MDM4). Conclusion: The six-IRL signature exhibits a promising prediction value of clinical prognosis and ICI efficacy in LUAD. Patients with low risk might gain benefits from ICI, although some have a risk of hyperprogressive disease.
Collapse
|
11
|
Balmert SC, Ghozloujeh ZG, Carey CD, Williams LH, Zhang J, Shahi P, Amer M, Sumpter TL, Erdos G, Korkmaz E, Falo LD. A microarray patch SARS-CoV-2 vaccine induces sustained antibody responses and polyfunctional cellular immunity. iScience 2022; 25:105045. [PMID: 36062075 PMCID: PMC9425707 DOI: 10.1016/j.isci.2022.105045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/19/2022] [Accepted: 08/25/2022] [Indexed: 12/01/2022] Open
Abstract
Sustainable global immunization campaigns against COVID-19 and other emerging infectious diseases require effective, broadly deployable vaccines. Here, we report a dissolvable microarray patch (MAP) SARS-CoV-2 vaccine that targets the immunoresponsive skin microenvironment, enabling efficacious needle-free immunization. Multicomponent MAPs delivering both SARS-CoV-2 S1 subunit antigen and the TLR3 agonist Poly(I:C) induce robust antibody and cellular immune responses systemically and in the respiratory mucosa. MAP vaccine-induced antibodies bind S1 and the SARS-CoV-2 receptor-binding domain, efficiently neutralize the virus, and persist at high levels for more than a year. The MAP platform reduces systemic toxicity of the delivered adjuvant and maintains vaccine stability without refrigeration. When applied to human skin, MAP vaccines activate skin-derived migratory antigen-presenting cells, supporting the feasibility of human translation. Ultimately, this shelf-stable MAP vaccine improves immunogenicity and safety compared to traditional intramuscular vaccines and offers an attractive alternative for global immunization efforts against a range of infectious pathogens.
Collapse
Affiliation(s)
- Stephen C. Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Li’an H. Williams
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Preeti Shahi
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Maher Amer
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L. Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Louis D. Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
12
|
Sparks R, Lau WW, Liu C, Han KL, Vrindten KL, Sun G, Cox M, Andrews SF, Bansal N, Failla LE, Manischewitz J, Grubbs G, King LR, Koroleva G, Leimenstoll S, Snow L, Chen J, Tang J, Mukherjee A, Sellers BA, Apps R, McDermott AB, Martins AJ, Bloch EM, Golding H, Khurana S, Tsang JS. Influenza vaccination and single cell multiomics reveal sex dimorphic immune imprints of prior mild COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.02.17.22271138. [PMID: 35233581 PMCID: PMC8887138 DOI: 10.1101/2022.02.17.22271138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Viral infections can have profound and durable functional impacts on the immune system. There is an urgent need to characterize the long-term immune effects of SARS-CoV-2 infection given the persistence of symptoms in some individuals and the continued threat of novel variants. Here we use systems immunology, including longitudinal multimodal single cell analysis (surface proteins, transcriptome, and V(D)J sequences) from 33 previously healthy individuals after recovery from mild, non-hospitalized COVID-19 and 40 age- and sex-matched healthy controls with no history of COVID-19 to comparatively assess the post-infection immune status (mean: 151 days after diagnosis) and subsequent innate and adaptive responses to seasonal influenza vaccination. Identification of both sex-specific and -independent temporally stable changes, including signatures of T-cell activation and repression of innate defense/immune receptor genes (e.g., Toll-like receptors) in monocytes, suggest that mild COVID-19 can establish new post-recovery immunological set-points. COVID-19-recovered males had higher innate, influenza-specific plasmablast, and antibody responses after vaccination compared to healthy males and COVID-19-recovered females, partly attributable to elevated pre-vaccination frequencies of a GPR56 expressing CD8+ T-cell subset in male recoverees that are "poised" to produce higher levels of IFNγ upon inflammatory stimulation. Intriguingly, by day 1 post-vaccination in COVID-19-recovered subjects, the expression of the repressed genes in monocytes increased and moved towards the pre-vaccination baseline of healthy controls, suggesting that the acute inflammation induced by vaccination could partly reset the immune states established by mild COVID-19. Our study reveals sex-dimorphic immune imprints and in vivo functional impacts of mild COVID-19 in humans, suggesting that prior COVID-19, and possibly respiratory viral infections in general, could change future responses to vaccination and in turn, vaccines could help reset the immune system after COVID-19, both in an antigen-agnostic manner.
Collapse
Affiliation(s)
- Rachel Sparks
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,These authors contributed equally
| | - William W. Lau
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,These authors contributed equally
| | - Can Liu
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,Graduate Program in Biological Sciences, University of Maryland, College Park, MD, USA,These authors contributed equally
| | - Kyu Lee Han
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Kiera L. Vrindten
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Guangping Sun
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Milann Cox
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | | | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Laura E. Failla
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Jody Manischewitz
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Lisa R. King
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Galina Koroleva
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - LaQuita Snow
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | | | - Jinguo Chen
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | | | | | - Richard Apps
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - Andrew J. Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Evan M. Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - John S. Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA,Correspondence:
| |
Collapse
|
13
|
Novak A, Pennings JLA, van der Maas L, Meiring HD, Ludwig I, Verkoeijen S, Rutten V, Broere F, Sloots A. Transcriptome and proteome analysis of innate immune responses to inactivated Leptospira and bivalent Leptospira vaccines in canine 030-D cells. Sci Rep 2022; 12:13418. [PMID: 35927283 PMCID: PMC9352656 DOI: 10.1038/s41598-022-16457-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/11/2022] [Indexed: 11/09/2022] Open
Abstract
Mandatory potency testing of Leptospira vaccine batches relies partially on in vivo procedures, requiring large numbers of laboratory animals. Cell-based assays could replace in vivo tests for vaccine quality control if biomarkers indicative of Leptospira vaccine potency are identified. We investigated innate immune responsiveness induced by inactivated L. interrogans serogroups Canicola and Icterohaemorrhagiae, and two bivalent, non-adjuvanted canine Leptospira vaccines containing the same serogroups. First, the transcriptome and proteome analysis of a canine monocyte/macrophage 030-D cell line stimulated with Leptospira strains, and vaccine B revealed more than 900 DEGs and 23 DEPs in common to these three stimuli. Second, comparison of responses induced by vaccine B and vaccine D revealed a large overlap in DEGs and DEPs as well, suggesting potential to identify biomarkers indicative of Leptospira vaccine quality. Because not many common DEPs were identified, we selected seven molecules from the identified DEGs, associated with pathways related to innate immunity, of which CXCL-10, IL-1β, SAA, and complement C3 showed increased secretion upon stimulation with both Leptospira vaccines. These molecules could be interesting targets for development of biomarker-based assays for Leptospira vaccine quality control in the future. Additionally, this study contributes to the understanding of the mechanisms by which Leptospira vaccines induce innate immune responses in the dog.
Collapse
Affiliation(s)
- Andreja Novak
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Intravacc, Bilthoven, The Netherlands
| | - Jeroen L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | | | - Irene Ludwig
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Saertje Verkoeijen
- Research Centre Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences Utrecht, Utrecht, The Netherlands
| | - Victor Rutten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Femke Broere
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Division of Internal Medicine of Companion Animals, Department of Clinical Science, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
14
|
Che J, Sun L, Shan J, Shi Y, Zhou Q, Zhao Y, Sun L. Artificial Lipids and Macrophage Membranes Coassembled Biomimetic Nanovesicles for Antibacterial Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201280. [PMID: 35616035 DOI: 10.1002/smll.202201280] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/23/2022] [Indexed: 06/15/2023]
Abstract
Tissue bacterial infections are a major pathological factor in many diseases. Effects on this aspect are in focus for the development of coordinated therapeutic strategies for bacterial killing and anti-inflammation. Here, inspired by the biodetoxification capacity of immune cells, multifunctional biomimetic nanovesicles (MϕM-LPs) that are co-assembled by macrophage membranes and artificial lipids to deliver antibiotics for treating bacterial infections, are presented. The macrophage membrane endows the MϕM-LPs with the capacity of lipopolysaccharide and inflammatory cytokine neutralization, while the artificial lipid membrane can be further engineered to increase the fluidity and anchor to bacteria. In addition, the MϕM-LPs can deliver sufficient ciprofloxacin with controllable release to accomplish an excellent antibacterial activity and biodetoxification capacity in vitro. Based on these advantages, it is demonstrated in a mouse model of Staphylococcus aureus (S. aureus) focal infection, that a single injection of the biomimetic nanovesicles can effectively anchor to and eliminate S. aureus in the infected tissue and reduce inflammatory cytokine levels. Thus, the tissue regeneration and collagen deposition can be accelerated. These results indicate the potential values of integrating natural and artificial membrane materials as a multifunctional biomimetic drug delivery system to treat bacterial infections.
Collapse
Affiliation(s)
- Junyi Che
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Lingyu Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingyang Shan
- Department of Neurology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518000, China
| | - Yong Shi
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Qing Zhou
- Department of Cardio-Thoracic Surgery, Institute of Translational Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
15
|
Huang J, Zhou X, Wang W, Zhou G, Zhang W, Gao Z, Wu X, Liu W. Combined analyses of RNA-sequence and Hi-C along with GWAS loci—A novel approach to dissect keloid disorder genetic mechanism. PLoS Genet 2022; 18:e1010168. [PMID: 35709140 PMCID: PMC9202908 DOI: 10.1371/journal.pgen.1010168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/25/2022] [Indexed: 12/05/2022] Open
Abstract
Keloid disorder is a tumour-like disease with invasive growth and a high recurrence rate. Genetic contribution is well expected due to the presence of autosomal dominant inheritance and various genetic mutations in keloid lesions. However, GWAS failed to reveal functional variants in exon regions but single nucleotide polymorphisms in the non-coding regions, suggesting the necessity of innovative genetic investigation. This study employed combined GWAS, RNA-sequence and Hi-C analyses to dissect keloid disorder genetic mechanisms using paired keloid tissues and normal skins. Differentially expressed genes, miRNAs and lncRNAs mined by RNA-sequence were identified to construct a network. From which, 8 significant pathways involved in keloid disorder pathogenesis were enriched and 6 of them were verified. Furthermore, topologically associated domains at susceptible loci were located via the Hi-C database and ten differentially expressed RNAs were identified. Among them, the functions of six molecules for cell proliferation, cell cycle and apoptosis were particularly examined and confirmed by overexpressing and knocking-down assays. This study firstly revealed unknown key biomarkers and pathways in keloid lesions using RNA-sequence and previously reported mutation loci, indicating a feasible approach to reveal the genetic contribution to keloid disorder and possibly to other diseases that are failed by GWAS analysis alone. Keloid disorder is a benign skin tumour characterized by uncontrolled fibroproliferative tissue growth, which only occurs in human beings with severe reoccurrence post-therapy. It affects several hundred million people with difficulty to control its growth and relapse. It has been long thought that exonic gene mutations must play an important role, but large-scaled GWAS analyses only revealed 3 single nucleotide polymorphisms in the non-coding regions as previously reported. For the first time, this study demonstrated that the true genetic mechanism is likely to be the dysfunctional epigenetic regulation caused by mutations in regulatory elements at the non-coding region as revealed by the combined analyses of GWAS, RNA-sequence and Hi-C data. This approach may lead to the breakthrough of keloid disorder genetic/epigenetic mechanism, if further large-scaled analyses are performed along with human keloid tissue Hi-C data.
Collapse
Affiliation(s)
- Jia Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering Research, Shanghai, China
| | - Xiaobo Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering Research, Shanghai, China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering Research, Shanghai, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering Research, Shanghai, China
- National Tissue Engineering Centre of China, Shanghai, China
| | - WenJie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering Research, Shanghai, China
- National Tissue Engineering Centre of China, Shanghai, China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoli Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering Research, Shanghai, China
- National Tissue Engineering Centre of China, Shanghai, China
- * E-mail:
| |
Collapse
|
16
|
Johnson-Weaver BT, Choi HW, Yang H, Granek JA, Chan C, Abraham SN, Staats HF. Nasal Immunization With Small Molecule Mast Cell Activators Enhance Immunity to Co-Administered Subunit Immunogens. Front Immunol 2021; 12:730346. [PMID: 34566991 PMCID: PMC8461742 DOI: 10.3389/fimmu.2021.730346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/23/2021] [Indexed: 01/02/2023] Open
Abstract
Mast cell activators are a novel class of mucosal vaccine adjuvants. The polymeric compound, Compound 48/80 (C48/80), and cationic peptide, Mastoparan 7 (M7) are mast cell activators that provide adjuvant activity when administered by the nasal route. However, small molecule mast cell activators may be a more cost-efficient adjuvant alternative that is easily synthesized with high purity compared to M7 or C48/80. To identify novel mast cell activating compounds that could be evaluated for mucosal vaccine adjuvant activity, we employed high-throughput screening to assess over 55,000 small molecules for mast cell degranulation activity. Fifteen mast cell activating compounds were down-selected to five compounds based on in vitro immune activation activities including cytokine production and cellular cytotoxicity, synthesis feasibility, and selection for functional diversity. These small molecule mast cell activators were evaluated for in vivo adjuvant activity and induction of protective immunity against West Nile Virus infection in BALB/c mice when combined with West Nile Virus envelope domain III (EDIII) protein in a nasal vaccine. We found that three of the five mast cell activators, ST101036, ST048871, and R529877, evoked high levels of EDIII-specific antibody and conferred comparable levels of protection against WNV challenge. The level of protection provided by these small molecule mast cell activators was comparable to the protection evoked by M7 (67%) but markedly higher than the levels seen with mice immunized with EDIII alone (no adjuvant 33%). Thus, novel small molecule mast cell activators identified by high throughput screening are as efficacious as previously described mast cell activators when used as nasal vaccine adjuvants and represent next-generation mast cell activators for evaluation in mucosal vaccine studies.
Collapse
Affiliation(s)
| | - Hae Woong Choi
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
| | - Hang Yang
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Josh A. Granek
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Cliburn Chan
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Soman N. Abraham
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
- Department of Immunology, School of Medicine, Duke University, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Herman F. Staats
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
- Department of Immunology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| |
Collapse
|
17
|
Pastor-Ibáñez R, Díez-Fuertes F, Sánchez-Palomino S, Alcamí J, Plana M, Torrents D, Leal L, García F. Impact of Transcriptome and Gut Microbiome on the Response of HIV-1 Infected Individuals to a Dendritic Cell-Based HIV Therapeutic Vaccine. Vaccines (Basel) 2021; 9:vaccines9070694. [PMID: 34202658 PMCID: PMC8310021 DOI: 10.3390/vaccines9070694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 01/08/2023] Open
Abstract
Therapeutic vaccines based on dendritic cells offer a good approach to HIV-specific T-cell responses and partial control of the viral load after antiretroviral therapy interruption. The aim of the present study was to identify mRNA expression profiles and to assess the impact of the gut microbiome composition for predicting the viral load control after antiretroviral therapy interruption. We enrolled 29 patients to receive either placebo or a monocyte-derived dendritic cell vaccine. Patients with a decrease in their viral load of >0.5 log10 copies/mL by 12 weeks after antiretroviral therapy interruption were considered responders. In total, 66 genes were considered differentially expressed between responders and non-responders. Enrichment analysis revealed several upregulated pathways involved in the host defense response to a virus via the type I interferon signaling pathway. Regarding the gut microbiota, responders showed enriched levels of Bacteroidetes (p < 0.005) and Verrucomicrobia (p = 0.017), while non-responders were enriched with Tenericutes (p = 0.049) and Actinobacteria (p < 0.005). We also found important differences at the genus level. However, we did not discover any effect of the dendritic cell vaccine on the transcriptome or the gut microbiota. An alternative analysis did characterize that the microbiota from responders were associated with the metabolic production of short-chain fatty acids, which are key metabolites in the regulation of intestinal homeostasis. The evidence now consistently shows that short-chain fatty acid depletion occurs in HIV-infected individuals receiving antiretroviral treatment.
Collapse
Affiliation(s)
- Roque Pastor-Ibáñez
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 170, 08036 Barcelona, Spain; (R.P.-I.); (S.S.-P.); (J.A.); (M.P.); (F.G.)
| | | | - Sonsoles Sánchez-Palomino
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 170, 08036 Barcelona, Spain; (R.P.-I.); (S.S.-P.); (J.A.); (M.P.); (F.G.)
| | - Jose Alcamí
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 170, 08036 Barcelona, Spain; (R.P.-I.); (S.S.-P.); (J.A.); (M.P.); (F.G.)
- Instituto de Salud Carlos III, Ctra. de Pozuelo, 28, Majadahonda, 28222 Madrid, Spain;
| | - Montserrat Plana
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 170, 08036 Barcelona, Spain; (R.P.-I.); (S.S.-P.); (J.A.); (M.P.); (F.G.)
| | - David Torrents
- Computational Genomics Groups, Barcelona Supercomputing Center (BSC), Plaça d’Eusebi Güell, 1-3, 08034 Barcelona, Spain;
| | - Lorna Leal
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 170, 08036 Barcelona, Spain; (R.P.-I.); (S.S.-P.); (J.A.); (M.P.); (F.G.)
- Infectious Diseases—Department, Hospital Clínic, IDIBAPS, University of Barcelona, Villarroel, 170, 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-932-275-586; Fax: +34-934-514-438
| | - Felipe García
- AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 170, 08036 Barcelona, Spain; (R.P.-I.); (S.S.-P.); (J.A.); (M.P.); (F.G.)
- Infectious Diseases—Department, Hospital Clínic, IDIBAPS, University of Barcelona, Villarroel, 170, 08036 Barcelona, Spain
| |
Collapse
|
18
|
d'Arminio Monforte A, Tavelli A, Perrone PM, Za A, Razzini K, Tomasoni D, Bordoni V, Romanò L, Orfeo N, Marchetti G, Colosio C. Association between previous infection with SARS CoV-2 and the risk of self-reported symptoms after mRNA BNT162b2 vaccination: Data from 3,078 health care workers. EClinicalMedicine 2021; 36:100914. [PMID: 34095793 PMCID: PMC8165130 DOI: 10.1016/j.eclinm.2021.100914] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Health care workers (HCWs) are at high risk of contracting an infection by SARS CoV-2 and thus they are a priority for vaccination. We hereby aim to investigate whether the risk of severe and moderate systemic symptoms (MSS) after vaccination is higher in HCWs with a history of previous COVID-19. METHODS An online questionnaire was offered to the cohort all HCWs undergoing anti-SARS CoV-2 mRNA BNT162b2 vaccination between January 4th and February 9th 2021 in two large tertiary hospitals (ASST Santi Paolo and Carlo) in Milan, Italy. Previous SARS-CoV-2 infection/COVID-19 was recorded. Local and systemic symptoms after each of the two doses were reported. MSS were those either interfering with daily activities or resulting in time off-work. Factors associated to MSS were identified by logistic regression. FINDINGS 3,078 HCW were included. Previous SARS-CoV-2 infection/COVID-19 occurred in 396 subjects (12·9%). 59·6% suffered from ≥1 local or systemic symptom after the first and 73·4% after the second dose. MSS occurred in 6·3% of cases (14·4% with previous vs 5·1% with no COVID-19 p<0·001) and in 28·3% (24·5% in COVID-19 vs 28·3% no COVID, p = 0·074) after the first and second dose, respectively. Subjects already experiencing COVID-19 had an independent 3-fold higher risk of MSS after the first and a 30% lower risk after the second dose. No severe adverse events were reported. INTERPRETATION Our data confirm in a real-world setting, the lack of severe adverse events and the short duration of reactogenicity in already infected HCWs. Possible differences in immune reactivity are drivers of MSS among this group of HCWs, as well as among females and younger individuals. FUNDING None.
Collapse
Affiliation(s)
- Antonella d'Arminio Monforte
- Unit of Infectious Diseases, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
- Corresponding author at: Unit of Infectious and Tropical Diseases, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Via A di Rudinì 8 -20142, Milan, Italy.
| | - Alessandro Tavelli
- Unit of Infectious Diseases, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Pier Mario Perrone
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Alessandro Za
- Medical Direction, ASST Santi Paolo e Carlo, Milan, Italy
| | - Katia Razzini
- Medical Direction, ASST Santi Paolo e Carlo, Milan, Italy
| | - Daniele Tomasoni
- Unit of Infectious Diseases, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Vittorio Bordoni
- Occupational Health Unit, International Centre for Rural Health, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Luisa Romanò
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Nicola Orfeo
- Medical Direction, ASST Santi Paolo e Carlo, Milan, Italy
| | - Giulia Marchetti
- Unit of Infectious Diseases, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Claudio Colosio
- Occupational Health Unit, International Centre for Rural Health, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| |
Collapse
|
19
|
Thiel N, Selwyn C, Murphy G, Simpson S, Chakrabarti AC. Recommendations for acceleration of vaccine development and emergency use filings for COVID-19 leveraging lessons from the novel oral polio vaccine. NPJ Vaccines 2021; 6:63. [PMID: 33888722 PMCID: PMC8062661 DOI: 10.1038/s41541-021-00325-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/18/2021] [Indexed: 11/17/2022] Open
Abstract
A new oral polio vaccine, nOPV2, has become the first vaccine to pursue a WHO Emergency Use Listing. Many lessons were learned as part of the accelerated development plan and submission, which have been categorized under the following sections: regulatory, clinical development, chemistry manufacturing and controls, and post-deployment monitoring. Efforts were made to adapt findings from these studies to COVID-19 vaccine candidates. Specific concepts for accelerating COVID-19 vaccine development across multiple functional domains were also included. The goals of this effort were twofold: (1) to help familiarize vaccine developers with the EUL process; and (2) to provide general guidance for faster development and preparations for launch during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Natalie Thiel
- University of Washington, School of Public Health, Seattle, WA, USA
| | - Casey Selwyn
- Bill & Melinda Gates Foundation, Seattle, WA, USA
| | | | | | | |
Collapse
|
20
|
Win Z, Weiner Rd J, Listanco A, Patel N, Sharma R, Greenwood A, Maertzdorf J, Mollenkopf HJ, Pizzoferro K, Cole T, Bodinham CL, Kaufmann SHE, Denoel P, Del Giudice G, Lewis DJM. Systematic Evaluation of Kinetics and Distribution of Muscle and Lymph Node Activation Measured by 18F-FDG- and 11C-PBR28-PET/CT Imaging, and Whole Blood and Muscle Transcriptomics After Immunization of Healthy Humans With Adjuvanted and Unadjuvanted Vaccines. Front Immunol 2021; 11:613496. [PMID: 33613536 PMCID: PMC7893084 DOI: 10.3389/fimmu.2020.613496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Systems vaccinology has been applied to detect signatures of human vaccine induced immunity but its ability, together with high definition in vivo clinical imaging is not established to predict vaccine reactogenicity. Within two European Commission funded high impact programs, BIOVACSAFE and ADITEC, we applied high resolution positron emission tomography/computed tomography (PET/CT) scanning using tissue-specific and non-specific radioligands together with transcriptomic analysis of muscle biopsies in a clinical model systematically and prospectively comparing vaccine-induced immune/inflammatory responses. 109 male participants received a single immunization with licensed preparations of either AS04-adjuvanted hepatitis B virus vaccine (AHBVV); MF59C-adjuvanted (ATIV) or unadjuvanted seasonal trivalent influenza vaccine (STIV); or alum-OMV-meningococcal B protein vaccine (4CMenB), followed by a PET/CT scan (n = 54) or an injection site muscle biopsy (n = 45). Characteristic kinetics was observed with a localized intramuscular focus associated with increased tissue glycolysis at the site of immunization detected by 18F-fluorodeoxyglucose (FDG) PET/CT, peaking after 1–3 days and strongest and most prolonged after 4CMenB, which correlated with clinical experience. Draining lymph node activation peaked between days 3–5 and was most prominent after ATIV. Well defined uptake of the immune cell-binding radioligand 11C-PBR28 was observed in muscle lesions and draining lymph nodes. Kinetics of muscle gene expression module upregulation reflected those seen previously in preclinical models with a very early (~6hrs) upregulation of monocyte-, TLR- and cytokine/chemokine-associated modules after AHBVV, in contrast to a response on day 3 after ATIV, which was bracketed by whole blood responses on day 1 as antigen presenting, inflammatory and innate immune cells trafficked to the site of immunization, and on day 5 associated with activated CD4+ T cells. These observations confirm the use of PET/CT, including potentially tissue-, cell-, or cytokine/chemokine-specific radioligands, is a safe and ethical quantitative technique to compare candidate vaccine formulations and could be safely combined with biopsy to guide efficient collection of samples for integrated whole blood and tissue systems vaccinology in small-scale but intensive human clinical models of immunization and to accelerate clinical development and optimisation of vaccine candidates, adjuvants, and formulations.
Collapse
Affiliation(s)
- Zarni Win
- Department of Nuclear Medicine and Radiological Sciences Unit, Imperial College Healthcare NHS Trust (ICHNT), London, United Kingdom
| | - January Weiner Rd
- Department for Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.,Core Unit for Bioinformatics (CUBI), Berlin Institute of Health, Berlin, Germany
| | - Allan Listanco
- National Institute for Health Research (NIHR) Imperial Clinical Research Facility (NICRF), Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Neva Patel
- Department of Nuclear Medicine and Radiological Sciences Unit, Imperial College Healthcare NHS Trust (ICHNT), London, United Kingdom
| | - Rohini Sharma
- Department of Surgery & Cancer, Imperial College London (ICL), London, United Kingdom
| | - Aldona Greenwood
- Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| | - Jeroen Maertzdorf
- Department for Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | - Kat Pizzoferro
- Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| | - Thomas Cole
- National Institute for Health Research (NIHR) Imperial Clinical Research Facility (NICRF), Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Caroline L Bodinham
- Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| | - Stefan H E Kaufmann
- Department for Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | | | - David J M Lewis
- National Institute for Health Research (NIHR) Imperial Clinical Research Facility (NICRF), Imperial College Healthcare NHS Trust, London, United Kingdom.,Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
21
|
Human Transcriptomic Response to the VSV-Vectored Ebola Vaccine. Vaccines (Basel) 2021; 9:vaccines9020067. [PMID: 33498214 PMCID: PMC7908976 DOI: 10.3390/vaccines9020067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Ebolavirus Disease (EVD) is a severe haemorrhagic fever that occurs in epidemic outbreaks, with a high fatality rate and no specific therapies available. rVSVΔG-ZEBOV-GP (Ervebo®), a live-attenuated recombinant vesicular stomatitis virus vector expressing the glycoprotein G of Zaire Ebolavirus, is the first vaccine approved for prevention of EVD. Both innate and adaptive responses are deemed to be involved in vaccine-induced protection, yet the mechanisms are not fully elucidated. A global transcriptomic approach was used to profile the blood host-response in 51 healthy volunteers enrolled in a phase 1/2 clinical trial. Signatures of the host responses were investigated assessing the enrichment in differentially expressed genes (DEGs) of specific “blood transcription modules” (BTM). Comparison of gene-expression levels showed that vaccination produces a peak of 5469 DEGs at day one, representing 38.6% of the expressed genes. Out of 346 BTMs, 144 were significantly affected by vaccination. Innate immunity pathways were induced from day 1 to day 14. At days 2 and 3, neutrophil modules were downregulated and complement-related modules upregulated. T-cell and cell-cycle associated modules were upregulated at days 7 and 14, while at day 28, no modules remained activated. At day 14, a direct correlation was observed between ZEBOV glycoprotein-specific antibody titres and activation of seven BTMs, including two related to B-cell activation and B cell receptor signalling. Transcriptomic analysis identified an rVSVΔG-ZEBOV-GP-induced signature and demonstrated a direct correlation of blood transcriptomic changes with ZEBOV glycoprotein-specific antibody titres.
Collapse
|
22
|
Hazlewood JE, Dumenil T, Le TT, Slonchak A, Kazakoff SH, Patch AM, Gray LA, Howley PM, Liu L, Hayball JD, Yan K, Rawle DJ, Prow NA, Suhrbier A. Injection site vaccinology of a recombinant vaccinia-based vector reveals diverse innate immune signatures. PLoS Pathog 2021; 17:e1009215. [PMID: 33439897 PMCID: PMC7837487 DOI: 10.1371/journal.ppat.1009215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/26/2021] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Poxvirus systems have been extensively used as vaccine vectors. Herein a RNA-Seq analysis of intramuscular injection sites provided detailed insights into host innate immune responses, as well as expression of vector and recombinant immunogen genes, after vaccination with a new multiplication defective, vaccinia-based vector, Sementis Copenhagen Vector. Chikungunya and Zika virus immunogen mRNA and protein expression was associated with necrosing skeletal muscle cells surrounded by mixed cellular infiltrates. The multiple adjuvant signatures at 12 hours post-vaccination were dominated by TLR3, 4 and 9, STING, MAVS, PKR and the inflammasome. Th1 cytokine signatures were dominated by IFNγ, TNF and IL1β, and chemokine signatures by CCL5 and CXCL12. Multiple signatures associated with dendritic cell stimulation were evident. By day seven, vaccine transcripts were absent, and cell death, neutrophil, macrophage and inflammation annotations had abated. No compelling arthritis signatures were identified. Such injection site vaccinology approaches should inform refinements in poxvirus-based vector design. Poxvirus vector systems have been widely developed for vaccine applications. Despite considerable progress, so far only one recombinant poxvirus vectored vaccine has to date been licensed for human use, with ongoing efforts seeking to enhance immunogenicity whilst minimizing reactogenicity. The latter two characteristics are often determined by early post-vaccination events at the injection site. We therefore undertook an injection site vaccinology approach to analyzing gene expression at the vaccination site after intramuscular inoculation with a recombinant, multiplication defective, vaccinia-based vaccine. This provided detailed insights into inter alia expression of vector-encoded immunoregulatory genes, as well as host innate and adaptive immune responses. We propose that such injection site vaccinology can inform rational vaccine vector design, and we discuss how the information and approach elucidated herein might be used to improve immunogenicity and limit reactogenicity of poxvirus-based vaccine vector systems.
Collapse
Affiliation(s)
- Jessamine E. Hazlewood
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Thuy T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andrii Slonchak
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Stephen H. Kazakoff
- Clinical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Ann-Marie Patch
- Clinical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Lesley-Ann Gray
- Australian Genome Research Facility Ltd., Melbourne, Australia
| | | | - Liang Liu
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - John D. Hayball
- Sementis Ltd., Hackney, Australia
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Daniel J. Rawle
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Natalie A. Prow
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Australian Infectious Disease Research Centre, Brisbane, Australia
- * E-mail:
| |
Collapse
|
23
|
Che J, Najer A, Blakney AK, McKay PF, Bellahcene M, Winter CW, Sintou A, Tang J, Keane TJ, Schneider MD, Shattock RJ, Sattler S, Stevens MM. Neutrophils Enable Local and Non-Invasive Liposome Delivery to Inflamed Skeletal Muscle and Ischemic Heart. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2003598. [PMID: 33103807 PMCID: PMC7613371 DOI: 10.1002/adma.202003598] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/03/2020] [Indexed: 05/24/2023]
Abstract
Uncontrolled inflammation is a major pathological factor underlying a range of diseases including autoimmune conditions, cardiovascular disease, and cancer. Improving localized delivery of immunosuppressive drugs to inflamed tissue in a non-invasive manner offers significant promise to reduce severe side effects caused by systemic administration. Here, a neutrophil-mediated delivery system able to transport drug-loaded nanocarriers to inflamed tissue by exploiting the inherent ability of neutrophils to migrate to inflammatory tissue is reported. This hybrid system (neutrophils loaded with liposomes ex vivo) efficiently migrates in vitro following an inflammatory chemokine gradient. Furthermore, the triggered release of loaded liposomes and reuptake by target macrophages is studied. The migratory behavior of liposome-loaded neutrophils is confirmed in vivo by demonstrating the delivery of drug-loaded liposomes to an inflamed skeletal muscle in mice. A single low-dose injection of the hybrid system locally reduces inflammatory cytokine levels. Biodistribution of liposome-loaded neutrophils in a human-disease-relevant myocardial ischemia reperfusion injury mouse model after i.v. injection confirms the ability of injected neutrophils to carry loaded liposomes to inflammation sites. This strategy shows the potential of nanocarrier-loaded neutrophils as a universal platform to deliver anti-inflammatory drugs to promote tissue regeneration in inflammatory diseases.
Collapse
Affiliation(s)
- Junyi Che
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Adrian Najer
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Anna K Blakney
- Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Paul F McKay
- Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Mohamed Bellahcene
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Charles W Winter
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Amalia Sintou
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Jiaqing Tang
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Timothy J Keane
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Michael D Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Robin J Shattock
- Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
24
|
Structural Basis for Vital Function and Malfunction of Serum Amyloid A: an Acute-Phase Protein that Wears Hydrophobicity on Its Sleeve. Curr Atheroscler Rep 2020; 22:69. [PMID: 32968930 PMCID: PMC7511256 DOI: 10.1007/s11883-020-00888-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review addresses normal and pathologic functions of serum amyloid A (SAA), an enigmatic biomarker of inflammation and protein precursor of AA amyloidosis, a life-threatening complication of chronic inflammation. SAA is a small, highly evolutionarily conserved acute-phase protein whose plasma levels increase up to one thousand-fold in inflammation, infection, or after trauma. The advantage of this dramatic but transient increase is unclear, and the complex role of SAA in immune response is intensely investigated. This review summarizes recent advances in our understanding of the structure-function relationship of this intrinsically disordered protein, outlines its newly emerging beneficial roles in lipid transport and inflammation control, and discusses factors that critically influence its misfolding in AA amyloidosis. RECENT FINDINGS High-resolution structures of lipid-free SAA in crystals and fibrils have been determined by x-ray crystallography and electron cryo-microscopy. Low-resolution structural studies of SAA-lipid complexes, together with biochemical, cell-based, animal model, genetic, and clinical studies, have provided surprising new insights into a wide range of SAA functions. An emerging vital role of SAA is lipid encapsulation to remove cell membrane debris from sites of injury. The structural basis for this role has been proposed. The lysosomal origin of AA amyloidosis has solidified, and its molecular and cellular mechanisms have emerged. Recent studies have revealed molecular underpinnings for understanding complex functions of this Cambrian protein in lipid transport, immune response, and amyloid formation. These findings help guide the search for much-needed targeted therapies to block the protein deposition in AA amyloidosis.
Collapse
|
25
|
Lauretti-Ferreira F, Silva PLD, Alcântara NM, Silva BF, Grabher I, Souza GO, Nakajima E, Akamatsu MA, Vasconcellos SA, Abreu PAE, Carvalho E, Martins EAL, Ho PL, da Silva JB. New strategies for Leptospira vaccine development based on LPS removal. PLoS One 2020; 15:e0230460. [PMID: 32218590 PMCID: PMC7100938 DOI: 10.1371/journal.pone.0230460] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/01/2020] [Indexed: 12/29/2022] Open
Abstract
Pathogenic spirochetes from genus Leptospira are etiologic agents of leptospirosis. Cellular vaccines against Leptospira infection often elicit mainly response against the LPS antigen of the serovars present in the formulation. There is no suitable protein candidate capable of replacing whole-cell vaccines, thus requiring new approaches on vaccine development to improve leptospirosis prevention. Our goal was to develop a whole-cell vaccine sorovar-independent based on LPS removal and conservation of protein antigens exposure, to evaluate the protective capacity of monovalent or bivalent vaccines against homologous and heterologous virulent Leptospira in hamster. Leptospire were subjected to heat inactivation, or to LPS extraction with butanol and in some cases further inactivation with formaldehyde. Hamsters were immunized and challenged with homologous or heterologous virulent serovars, blood and organs were collected from the survivors for bacterial quantification, chemokine evaluation, and analysis of sera antibody reactivity and cross-reactivity by Western blot. Immunization with either heated or low LPS vaccines with serovar Copenhageni or Canicola resulted in 100% protection of the animals challenged with homologous virulent bacteria. Notably, different from the whole-cell vaccine, the low LPS vaccines produced with serovar Canicola provided only partial protection in heterologous challenge with the virulent Copenhageni serovar. Immunization with bivalent formulation results in 100% protection of immunized animals challenged with virulent serovar Canicola. All vaccines produced were able to eliminate bacteria from the kidney of challenged animals. All the vaccines raised antibodies capable to recognize antigens of serovars not present in the vaccine formulation. Transcripts of IFNγ, CXCL16, CCL5, CXCL10, CXCR6, and CCR5, increased in all immunized animals. Conclusion: Our results showed that bivalent vaccines with reduced LPS may be an interesting strategy for protection against heterologous virulent serovars. Besides the desirable multivalent protection, the low LPS vaccines are specially promising due to the expected lower reatogenicity.
Collapse
Affiliation(s)
- Fabiana Lauretti-Ferreira
- Bioindustrial Division, Butantan Institute, São Paulo, Brazil
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | | | | | - Bruna F. Silva
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | - Isabele Grabher
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | - Gisele O. Souza
- Laboratory of Bacterial Zoonosis, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Erika Nakajima
- Laboratory of Process Development, Butantan Institute, São Paulo, Brazil
| | | | - Silvio A. Vasconcellos
- Laboratory of Bacterial Zoonosis, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Eneas Carvalho
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
| | | | - Paulo L. Ho
- Bioindustrial Division, Butantan Institute, São Paulo, Brazil
| | - Josefa B. da Silva
- Laboratory of Bacteriology, Butantan Institute, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
26
|
Tregoning JS, Weiner J, Cizmeci D, Hake D, Maertzdorf J, Kaufmann SHE, Leroux-Roels G, Maes C, Aerssens A, Calvert A, Jones CE. Pregnancy has a minimal impact on the acute transcriptional signature to vaccination. NPJ Vaccines 2020; 5:29. [PMID: 32219001 PMCID: PMC7096498 DOI: 10.1038/s41541-020-0177-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/24/2020] [Indexed: 01/15/2023] Open
Abstract
Vaccination in pregnancy is an effective tool to protect both the mother and infant; vaccines against influenza, pertussis and tetanus are currently recommended. A number of vaccines with a specific indication for use in pregnancy are in development, with the specific aim of providing passive humoral immunity to the newborn child against pathogens responsible for morbidity and mortality in young infants. However, the current understanding about the immune response to vaccination in pregnancy is incomplete. We analysed the effect of pregnancy on early transcriptional responses to vaccination. This type of systems vaccinology approach identifies genes and pathways that are altered in response to vaccination and can be used to understand both the acute inflammation in response to the vaccine and to predict immunogenicity. Pregnant women and mice were immunised with Boostrix-IPV, a multivalent vaccine, which contains three pertussis antigens. Blood was collected from women before and after vaccination and RNA extracted for analysis by microarray. While there were baseline differences between pregnant and non-pregnant women, vaccination induced characteristic patterns of gene expression, with upregulation in interferon response and innate immunity gene modules, independent of pregnancy. We saw similar patterns of responses in both women and mice, supporting the use of mice for preclinical screening of novel maternal vaccines. Using a systems vaccinology approach in pregnancy demonstrated that pregnancy does not affect the initial response to vaccination and that studies in non-pregnant women can provide information about vaccine immunogenicity and potentially safety.
Collapse
Affiliation(s)
- John S Tregoning
- 1Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG UK
| | - January Weiner
- 2Max Planck Institute for Infection Biology, Berlin, Germany.,6Present Address: Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Deniz Cizmeci
- 1Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG UK
| | - Danielle Hake
- 3Vaccine Institute, St George's, University of London, London, UK
| | | | | | - Geert Leroux-Roels
- 4Centre for Vaccinology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Cathy Maes
- 4Centre for Vaccinology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Annelies Aerssens
- 4Centre for Vaccinology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Anna Calvert
- 3Vaccine Institute, St George's, University of London, London, UK
| | - Christine E Jones
- 3Vaccine Institute, St George's, University of London, London, UK.,5Faculty of Medicine and Institute for Life Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
27
|
Characterization of potential biomarkers of reactogenicity of licensed antiviral vaccines: randomized controlled clinical trials conducted by the BIOVACSAFE consortium. Sci Rep 2019; 9:20362. [PMID: 31889148 PMCID: PMC6937244 DOI: 10.1038/s41598-019-56994-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/10/2019] [Indexed: 02/08/2023] Open
Abstract
Biomarkers predictive of inflammatory events post-vaccination could accelerate vaccine development. Within the BIOVACSAFE framework, we conducted three identically designed, placebo-controlled inpatient/outpatient clinical studies (NCT01765413/NCT01771354/NCT01771367). Six antiviral vaccination strategies were evaluated to generate training data-sets of pre-/post-vaccination vital signs, blood changes and whole-blood gene transcripts, and to identify putative biomarkers of early inflammation/reactogenicity that could guide the design of subsequent focused confirmatory studies. Healthy adults (N = 123; 20-21/group) received one immunization at Day (D)0. Alum-adjuvanted hepatitis B vaccine elicited vital signs and inflammatory (CRP/innate cells) responses that were similar between primed/naive vaccinees, and low-level gene responses. MF59-adjuvanted trivalent influenza vaccine (ATIV) induced distinct physiological (temperature/heart rate/reactogenicity) response-patterns not seen with non-adjuvanted TIV or with the other vaccines. ATIV also elicited robust early (D1) activation of IFN-related genes (associated with serum IP-10 levels) and innate-cell-related genes, and changes in monocyte/neutrophil/lymphocyte counts, while TIV elicited similar but lower responses. Due to viral replication kinetics, innate gene activation by live yellow-fever or varicella-zoster virus (YFV/VZV) vaccines was more suspended, with early IFN-associated responses in naïve YFV-vaccine recipients but not in primed VZV-vaccine recipients. Inflammatory responses (physiological/serum markers, innate-signaling transcripts) are therefore a function of the vaccine type/composition and presence/absence of immune memory. The data reported here have guided the design of confirmatory Phase IV trials using ATIV to provide tools to identify inflammatory or reactogenicity biomarkers.
Collapse
|
28
|
Chevrier N. Decoding the Body Language of Immunity: Tackling the Immune System at the Organism Level. ACTA ACUST UNITED AC 2019; 18:19-26. [PMID: 32490290 DOI: 10.1016/j.coisb.2019.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The immune system is a dynamic mesh of molecules, cells and tissues spanning the entire organism. Despite a wealth of knowledge about the components of the immune system, little is known about the general rules governing the organismal circuitry of immunity. Deciphering the immune system at the scale of the whole organism is crucial to understanding fundamental problems in immunobiology and physiology, and to manipulate immunity for maintaining health and preventing disease. Here I discuss the emerging principles of inter-organ communications during immune responses by focusing on three common themes that are the regulation of the (i) composition, (ii) condition and (iii) coordination of communicating organs by molecular and cellular factors. Based on these common principles, I emphasize fundamental gaps in our knowledge of organismal immune processes and the outlook to tackle immunity at the scale of the whole organism.
Collapse
Affiliation(s)
- Nicolas Chevrier
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|