1
|
Ijaz M, Hasan I, Chaudhry TH, Huang R, Zhang L, Hu Z, Tan Q, Guo B. Bacterial derivatives mediated drug delivery in cancer therapy: a new generation strategy. J Nanobiotechnology 2024; 22:510. [PMID: 39182109 PMCID: PMC11344338 DOI: 10.1186/s12951-024-02786-w] [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: 06/06/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024] Open
Abstract
Cancer is measured as a major threat to human life and is a leading cause of death. Millions of cancer patients die every year, although a burgeoning number of researchers have been making tremendous efforts to develop cancer medicine to fight against cancer. Owing to the complexity and heterogeneity of cancer, lack of ability to treat deep tumor tissues, and high toxicity to the normal cells, it complicates the therapy of cancer. However, bacterial derivative-mediated drug delivery has raised the interest of researchers in overcoming the restrictions of conventional cancer chemotherapy. In this review, we show various examples of tumor-targeting bacteria and bacterial derivatives for the delivery of anticancer drugs. This review also describes the advantages and limitations of delivering anticancer treatment drugs under regulated conditions employing these tumor-targeting bacteria and their membrane vesicles. This study highlights the substantial potential for clinical translation of bacterial-based drug carriers, improve their ability to work with other treatment modalities, and provide a more powerful, dependable, and distinctive tumor therapy.
Collapse
Affiliation(s)
- Muhammad Ijaz
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518060, Guangdong, China
| | - Tamoor Hamid Chaudhry
- Antimicrobial Resistance (AMR) Containment & Infection Prevention & Control (IPC) Program, National Institute of Health, Chak Shahzad, Islamabad, Pakistan
| | - Rui Huang
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Lan Zhang
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Ziwei Hu
- Institute of Otolaryngology Head and Neck Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, China.
| | - Qingqin Tan
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China.
| |
Collapse
|
2
|
Park SH, Song HK, Ji KY, Jung DH, Jang S, Kim T, Kim HK. Topical Administration of Gardenia jasminoides Extract Regulates Th2 Immunity in OVA-Induced Mice. Cells 2023; 12:cells12060941. [PMID: 36980282 PMCID: PMC10047210 DOI: 10.3390/cells12060941] [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: 02/14/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
A key feature of an allergic immune response is a T helper type 2 (Th2)-mediated response with production of allergen-specific IgE antibodies. Gardenia jasminoides extract with the crocin removed (GJExCR) has been shown to inhibit IgE-mediated allergic disease. To evaluate the efficacy and mechanism-of-action of this inhibition, GJExCR was used in an ovalbumin (OVA)-induced allergy model in BALB/C mice. Sensitization of BALB/C mice with OVA and aluminum hydroxide was performed on days 1 and 14 by intraperitoneal injection, followed by OVA challenge to the dorsal skin for 2 weeks before removal. Seven days post-challenge, mice were treated with GJExCR topically every day for 11 days. Enzyme-linked immunosorbent assay, flow cytometry analysis, real-time PCR, and western blot were performed to determine IgE and Th2 cytokine levels. Following OVA challenge, Th2 cytokine expression and both total and OVA-specific serum IgE levels increased, of which OVA-specific IgE and Th2 cytokine levels decreased after GJExCR treatment. Flow cytometry analysis revealed that GJExCR treatment decreased CD4+ and CD8+ T cell populations in the spleen and lymph nodes. In addition, treatment with GJExCR downregulated signal transducer and activator of transcription 1 (STAT1) activation and Th2 cytokine levels as compared to control. GJExCR containing geniposide downregulated STAT1 activation in HaCaT cells. These findings demonstrate that GJExCR exerts its anti-allergy effect via inhibition of STAT1 activation, thus regulating the immune response via modulation of Th2 cytokine release and IgE levels. Therefore, we propose GJExCR as a potential treatment for allergic hypersensitivity reactions.
Collapse
Affiliation(s)
- Sun Haeng Park
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Republic of Korea
| | - Hyun-Kyung Song
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Republic of Korea
| | - Kon-Young Ji
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Republic of Korea
| | - Dong Ho Jung
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Republic of Korea
| | - Seol Jang
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Republic of Korea
| | - Taesoo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Republic of Korea
| | - Ho Kyoung Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 305-811, Republic of Korea
| |
Collapse
|
3
|
Howell LM, Forbes NS. Bacteria-based immune therapies for cancer treatment. Semin Cancer Biol 2021; 86:1163-1178. [PMID: 34547442 DOI: 10.1016/j.semcancer.2021.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 12/23/2022]
Abstract
Engineered bacterial therapies that target the tumor immune landscape offer a new class of cancer immunotherapy. Salmonella enterica and Listeria monocytogenes are two species of bacteria that have been engineered to specifically target tumors and serve as delivery vessels for immunotherapies. Therapeutic bacteria have been engineered to deliver cytokines, gene silencing shRNA, and tumor associated antigens that increase immune activation. Bacterial therapies stimulate both the innate and adaptive immune system, change the immune dynamics of the tumor microenvironment, and offer unique strategies for targeting tumors. Bacteria have innate adjuvant properties, which enable both the delivered molecules and the bacteria themselves to stimulate immune responses. Bacterial immunotherapies that deliver cytokines and tumor-associated antigens have demonstrated clinical efficacy. Harnessing the diverse set of mechanisms that Salmonella and Listeria use to alter the tumor-immune landscape has the potential to generate many new and effective immunotherapies.
Collapse
Affiliation(s)
- Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States
| | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States.
| |
Collapse
|
4
|
Si Y, Tian Q, Zhao F, Kelly SH, Shores LS, Camacho DF, Sperling AI, Andrade MS, Collier JH, Chong AS. Adjuvant-free nanofiber vaccine induces in situ lung dendritic cell activation and T H17 responses. SCIENCE ADVANCES 2020; 6:eaba0995. [PMID: 32821819 PMCID: PMC7413739 DOI: 10.1126/sciadv.aba0995] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 06/25/2020] [Indexed: 05/07/2023]
Abstract
The current paradigm that subunit vaccines require adjuvants to optimally activate innate immunity implies that increased vaccine reactogenicity will invariably be linked to improved immunogenicity. Countering this paradigm, nanoparticulate vaccines have been reported to act as delivery systems for vaccine antigens and induce immunity without the need for exogenous adjuvants or local inflammation; however, the mechanisms underlying the immunogenicity of nanoparticle vaccines are incompletely identified. Here, we show that antigens displayed on self-assembling nanofiber scaffolds and delivered intranasally are presented by CD103+ and CD11b+ lung dendritic cells that up-regulate CD80 and migrate into the draining lymph node (LN). This was accompanied by a nearly exclusive priming and accumulation of antigen-specific TH17 cells occurring independently in both LN and lung. Thus, self-assembling peptide nanofiber vaccines may represent a novel, needle- and adjuvant-free means of eliciting protective immunity against fungal and bacterial infections at skin and mucosal barrier surfaces.
Collapse
Affiliation(s)
- Youhui Si
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiaomu Tian
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Fan Zhao
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Sean H. Kelly
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
| | - Lucas S. Shores
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
| | - Daniel F. Camacho
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Anne I. Sperling
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Michael S. Andrade
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Joel H. Collier
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
- Corresponding author. (A.S.C.); (J.H.C.)
| | - Anita S. Chong
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
- Corresponding author. (A.S.C.); (J.H.C.)
| |
Collapse
|
5
|
Troy A, Esparza-Gonzalez SC, Bartek A, Creissen E, Izzo L, Izzo AA. Pulmonary mucosal immunity mediated through CpG provides adequate protection against pulmonary Mycobacterium tuberculosis infection in the mouse model. A role for type I interferon. Tuberculosis (Edinb) 2020; 123:101949. [PMID: 32741537 DOI: 10.1016/j.tube.2020.101949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/28/2022]
Abstract
Toll-Like Receptor (TLR) 9 stimulation is required for induction of potent immune responses against pathogen invasion. The use of unmethylated CpG as adjuvants in vaccines provides an excellent means of stimulating adaptive immunity. Our data demonstrate that CpG-C provided prolonged immune responses in the mouse model of tuberculosis when formulated with liposomes and the Mycobacterium tuberculosis antigen ESAT-6. A reduction in the mycobacterial burden was best achieved when administered as an intranasal vaccine and was dependent on type I interferon (IFN). There was a significant difference between CpG-C inoculated wild type and IFN-αR1-/- mice, indicating that type I IFN plays a role in the immune response following CpG-C inoculation. Further analysis showed that early NK cell presence was not an absolute requirement, although elevated IFN-γ levels were detected in the lungs of mice within 48 h. The reduction in mycobacterial burden was MyD88-independent as CpG-C inoculated MyD88-/- mice showed comparable mycobacterial burdens to wild type mice with no detriment due to the lack of MyD88. Together our data show that pulmonary stimulation of TLR9 bearing antigen presenting cells resulted in the induction of protective immunity against M. tuberculosis infection that was dependent on type I IFN signaling.
Collapse
Affiliation(s)
- Amber Troy
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Sandra C Esparza-Gonzalez
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Alicia Bartek
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Elizabeth Creissen
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Linda Izzo
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Angelo A Izzo
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA.
| |
Collapse
|
6
|
Contact-dependent delivery of IL-2 by dendritic cells to CD4 T cells in the contraction phase promotes their long-term survival. Protein Cell 2019; 11:108-123. [PMID: 31691194 PMCID: PMC6954898 DOI: 10.1007/s13238-019-00662-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/18/2019] [Indexed: 01/17/2023] Open
Abstract
Common γ chain cytokines are important for immune memory formation. Among them, the role of IL-2 remains to be fully explored. It has been suggested that this cytokine is critically needed in the late phase of primary CD4 T cell activation. Lack of IL-2 at this stage sets for a diminished recall response in subsequent challenges. However, as IL-2 peak production is over at this point, the source and the exact mechanism that promotes its production remain elusive. We report here that resting, previously antigen-stimulated CD4 T cells maintain a minimalist response to dendritic cells after their peak activation in vitro. This subtle activation event may be induced by DCs without overt presence of antigen and appears to be stronger if IL-2 comes from the same dendritic cells. This encounter reactivates a miniature IL-2 production and leads a gene expression profile change in these previously activated CD4 T cells. The CD4 T cells so experienced show enhanced reactivation intensity upon secondary challenges later on. Although mostly relying on in vitro evidence, our work may implicate a subtle programing for CD4 T cell survival after primary activation in vivo.
Collapse
|
7
|
Recent trends and advances in microbe-based drug delivery systems. ACTA ACUST UNITED AC 2019; 27:799-809. [PMID: 31376116 DOI: 10.1007/s40199-019-00291-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
Since more than a decade, pharmaceutical researchers endeavor to develop an effective, safe and target-specific drug delivery system to potentiate the therapeutic actions and reduce the side effects. The conventional drug delivery systems (DDSs) show the improvement in the lifestyle of the patients suffering from non-communicable diseases, autoimmune diseases but sometimes, drug resistance developed during the treatment is a major concern for clinicians to find an alternative and more advanced transport systems. Advancements in drug delivery facilitate the development of active carrier for targeted action with improved pharmacokinetic behavior. This review article focuses on microbe-based drug delivery systems to provide safe, non-toxic, site-specific targeted action with lesser side effects. Pharmaceutical researchers play a vital part in microbe-based drug delivery systems as a therapeutic agent and carrier. The properties of microorganisms like self-propulsion, in-situ production of therapeutics, penetration into the tumor cells, increase in immunity, etc. are of interest for development of highly effective delivery carrier. Lactococcus lactis is therapeutically helpful in Inflammatory Bowel Disease (IBD) and is under investigation of phase I clinical trial. Moreover, bacteria, anti-cancer oncolytic viruses, viral vectors (gene therapy) and viral immunotherapy are the attractive areas of biotechnological research. Virus acts as a distinctive candidate for imaging of tumor and accumulation of active in tumor. Graphical abstract Classification of microbe-based drug delivery system.
Collapse
|
8
|
Namvarpour M, Tebianian M, Mansouri R, Ebrahimi SM, Kashkooli S. Comparison of different immunization routes on the immune responses induced by Mycobacterium tuberculosis ESAT-6/CFP-10 recombinant protein. Biologicals 2019; 59:6-11. [PMID: 31014910 DOI: 10.1016/j.biologicals.2019.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 12/20/2018] [Accepted: 04/14/2019] [Indexed: 01/09/2023] Open
Abstract
According to some difficulties against tuberculosis (TB) vaccination, development of new TB vaccines has been noted in recent years. Selection of proper route for vaccination is one of the most important factors for induction of good immune responses. Hence, in this study, the effects of different administration routes, including intranasal (I.N), subcutaneous (S.C) and intramuscular (I.M) on immune responses against Mycobacterium tuberculosis ESAT-6/CFP-10 recombinant protein has been considered. Recombinant ESAT-6/CFP-10 protein with or without adjuvant (MF59 or cholera toxin B (CTB)) was administered by three routes of I.M, I.N and S.C to mice for three times. Then, the levels of specific antibodies, lymphocyte proliferation and IFN-γ/IL-5 cytokine profile have been carried out to evaluate the humoral and cellular responses. The results showed that the titers of specific antibodies were quickly elevated in S.C and I.M groups after first immunization. Otherwise, the raise of antibody has delay in the I.N immunized animals. The levels of IFN-γ and lymphocyte proliferation have been increased in all of vaccinated groups. However, the I.N immunized mice have lower levels of IL-5 production. Based on our finding, the ESAT-6/CFP-10 recombinant protein is a potent stimulator of immune responses in all of three immunization strategies. However intranasal administration of this antigen has tended to reinforcement of cellular immune responses.
Collapse
Affiliation(s)
- Mozhdeh Namvarpour
- Department of Immunology, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Majid Tebianian
- Department of Biotechnology, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran.
| | - Reza Mansouri
- Department of Immunology, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Seyyed Mahmoud Ebrahimi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, P.O. Box 14155-3651, Tehran, Iran
| | - Shiva Kashkooli
- - Department of Biotechnology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, (IAUPS), Tehran, Iran
| |
Collapse
|
9
|
Santoro F, Pettini E, Kazmin D, Ciabattini A, Fiorino F, Gilfillan GD, Evenroed IM, Andersen P, Pozzi G, Medaglini D. Transcriptomics of the Vaccine Immune Response: Priming With Adjuvant Modulates Recall Innate Responses After Boosting. Front Immunol 2018; 9:1248. [PMID: 29922291 PMCID: PMC5996052 DOI: 10.3389/fimmu.2018.01248] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/18/2018] [Indexed: 12/17/2022] Open
Abstract
Transcriptomic profiling of the immune response induced by vaccine adjuvants is of critical importance for the rational design of vaccination strategies. In this study, transcriptomics was employed to profile the effect of the vaccine adjuvant used for priming on the immune response following re-exposure to the vaccine antigen alone. Mice were primed with the chimeric vaccine antigen H56 of Mycobacterium tuberculosis administered alone or with the CAF01 adjuvant and boosted with the antigen alone. mRNA sequencing was performed on blood samples collected 1, 2, and 7 days after priming and after boosting. Gene expression analysis at day 2 after priming showed that the CAF01 adjuvanted vaccine induced a stronger upregulation of the innate immunity modules compared with the unadjuvanted formulation. The immunostimulant effect of the CAF01 adjuvant, used in the primary immunization, was clearly seen after a booster immunization with a low dose of antigen alone. One day after boost, we observed a strong upregulation of multiple genes in blood of mice primed with H56 + CAF01 compared with mice primed with the H56 alone. In particular, blood transcription modules related to innate immune response, such as monocyte and neutrophil recruitment, activation of antigen-presenting cells, and interferon response were activated. Seven days after boost, differential expression of innate response genes faded while a moderate differential expression of T cell activation modules was appreciable. Indeed, immunological analysis showed a higher frequency of H56-specific CD4+ T cells and germinal center B cells in draining lymph nodes, a strong H56-specific humoral response and a higher frequency of antibody-secreting cells in spleen of mice primed with H56 + CAF01. Taken together, these data indicate that the adjuvant used for priming strongly reprograms the immune response that, upon boosting, results in a stronger recall innate response essential for shaping the downstream adaptive response.
Collapse
Affiliation(s)
- Francesco Santoro
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Dmitri Kazmin
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Gregor D Gilfillan
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Ida M Evenroed
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Gianni Pozzi
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| |
Collapse
|
10
|
Ciabattini A, Pettini E, Fiorino F, Lucchesi S, Pastore G, Brunetti J, Santoro F, Andersen P, Bracci L, Pozzi G, Medaglini D. Heterologous Prime-Boost Combinations Highlight the Crucial Role of Adjuvant in Priming the Immune System. Front Immunol 2018; 9:380. [PMID: 29593710 PMCID: PMC5857569 DOI: 10.3389/fimmu.2018.00380] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/12/2018] [Indexed: 12/21/2022] Open
Abstract
The induction and modulation of the immune response to vaccination can be rationally designed by combining different vaccine formulations for priming and boosting. Here, we investigated the impact of heterologous prime-boost approaches on the vaccine-specific cellular and humoral responses specific for a mycobacterial vaccine antigen. C57BL/6 mice were primed with the chimeric vaccine antigen H56 administered alone or with the CAF01 adjuvant, and boosted with H56 alone, or combined with CAF01 or with the squalene-based oil-in-water emulsion adjuvant (o/w squalene). A strong secondary H56-specific CD4+ T cell response was recalled by all the booster vaccine formulations when mice had been primed with H56 and CAF01, but not with H56 alone. The polyfunctional nature of T helper cells was analyzed and visualized with the multidimensional flow cytometry FlowSOM software, implemented as a package of the R environment. A similar cytokine profile was detected in groups primed with H56 + CAF01 and boosted with or without adjuvant, except for some clusters of cells expressing high level of IL-17 together with TNF-α, IL-2, and IFN-γ, that were significantly upregulated only in groups boosted with the adjuvants. On the contrary, the comparison between groups primed with or without the adjuvant showed a completely different clusterization of cells, strengthening the impact of the formulation used for primary immunization on the profiling of responding cells. The presence of the CAF01 adjuvant in the priming formulation deeply affected also the secondary humoral response, especially in groups boosted with H56 alone or o/w squalene. In conclusion, the presence of CAF01 adjuvant in the primary immunization is crucial for promoting primary T and B cell responses that can be efficiently reactivated by booster immunization also performed with antigen alone.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Jlenia Brunetti
- U&E PreMed Laboratory, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Luisa Bracci
- U&E PreMed Laboratory, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
11
|
Kalam MA, Khan AA, Alshamsan A. Non-invasive administration of biodegradable nano-carrier vaccines. Am J Transl Res 2017; 9:15-35. [PMID: 28123631 PMCID: PMC5250701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/29/2016] [Indexed: 06/06/2023]
Abstract
Polymeric nanoparticulate carriers play an important role and holding a significant potential for the development of novel immunomodulatory agents as easily they are taken up by antigen presenting cells. They allow an enhanced antigen stability, better immunogenicity and immunostimulatory effect with sustained and controlled release of the antigen to the target sites. Better information and vital understanding of mechanism of action, interaction of such vectors with the APCs and dendritic cells and antigen release kinetics in immunomodulatory effects, and improved knowledge of their in vivo fate and distribution are now needed, those collectively would speed up the rational strategies of nanoparticles as carriers for vaccines and other protein antigens. The evolution of such innovative adjuvants for protein and DNA immunizations are an exciting and growing zone in immunology, which may enhance the clinical outcomes in many infectious and non-infectious diseases. This review summarizes the recent advances in nano-vaccinology with polymeric (especially biodegradable) carriers, their methods of preparation, surface modification, their interaction with antigen presenting cells, release of antigens, its kinetics and mechanism in the delivery of vaccines via non-invasive routes.
Collapse
Affiliation(s)
- Mohd Abul Kalam
- Nanomedicine Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud UniversityP. O. Box: 2457, Riyadh 11451, Saudi Arabia
| | - Abdul Arif Khan
- Nanomedicine Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud UniversityP. O. Box: 2457, Riyadh 11451, Saudi Arabia
| | - Aws Alshamsan
- Nanomedicine Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud UniversityP. O. Box: 2457, Riyadh 11451, Saudi Arabia
- King Abdullah Institute for Nanotechnology, King Saud UniversityP. O. Box: 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
12
|
Bioengineered and biohybrid bacteria-based systems for drug delivery. Adv Drug Deliv Rev 2016; 106:27-44. [PMID: 27641944 DOI: 10.1016/j.addr.2016.09.007] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
The use of bacterial cells as agents of medical therapy has a long history. Research that was ignited over a century ago with the accidental infection of cancer patients has matured into a platform technology that offers the promise of opening up new potential frontiers in medical treatment. Bacterial cells exhibit unique characteristics that make them well-suited as smart drug delivery agents. Our ability to genetically manipulate the molecular machinery of these cells enables the customization of their therapeutic action as well as its precise tuning and spatio-temporal control, allowing for the design of unique, complex therapeutic functions, unmatched by current drug delivery systems. Early results have been promising, but there are still many important challenges that must be addressed. We present a review of promises and challenges of employing bioengineered bacteria in drug delivery systems and introduce the biohybrid design concept as a new additional paradigm in bacteria-based drug delivery.
Collapse
|
13
|
Ciabattini A, Pettini E, Fiorino F, Pastore G, Andersen P, Pozzi G, Medaglini D. Modulation of Primary Immune Response by Different Vaccine Adjuvants. Front Immunol 2016; 7:427. [PMID: 27781036 PMCID: PMC5066114 DOI: 10.3389/fimmu.2016.00427] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/28/2016] [Indexed: 01/09/2023] Open
Abstract
Adjuvants contribute to enhancing and shaping the vaccine immune response through different modes of action. Here early biomarkers of adjuvanticity after primary immunization were investigated using four different adjuvants combined with the chimeric tuberculosis vaccine antigen H56. C57BL/6 mice were immunized by the subcutaneous route with different vaccine formulations, and the modulation of primary CD4+ T cell and B cell responses was assessed within draining lymph nodes, blood, and spleen, 7 and 12 days after priming. Vaccine formulations containing the liposome system CAF01 or a squalene-based oil-in-water emulsion (o/w squalene), but not aluminum hydroxide (alum) or CpG ODN 1826, elicited a significant primary antigen-specific CD4+ T cell response compared to antigen alone, 7 days after immunization. The effector function of activated CD4+ T cells was skewed toward a Th1/Th17 response by CAF01, while a Th1/Th2 response was elicited by o/w squalene. Differentiation of B cells in short-lived plasma cells, and subsequent early H56-specific IgG secretion, was observed in mice immunized with o/w squalene or CpG adjuvants. Tested adjuvants promoted the germinal center reaction with different magnitude. These results show that the immunological activity of different adjuvants can be characterized by profiling early immunization biomarkers after primary immunization. These data and this approach could give an important contribution to the rational development of heterologous prime–boost vaccine immunization protocols.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut , Copenhagen , Denmark
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| |
Collapse
|
14
|
Zorgi NE, Galisteo AJ, Sato MN, do Nascimento N, de Andrade HF. Immunity in the spleen and blood of mice immunized with irradiated Toxoplasma gondii tachyzoites. Med Microbiol Immunol 2016; 205:297-314. [PMID: 26732075 DOI: 10.1007/s00430-015-0447-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/21/2015] [Indexed: 01/14/2023]
Abstract
Toxoplasma gondii infection induces a strong and long-lasting immune response that is able to prevent most reinfections but allows tissue cysts. Irradiated, sterilized T. gondii tachyzoites are an interesting vaccine, and they induce immunity that is similar to infection, but without cysts. In this study, we evaluated the cellular immune response in the blood and spleen of mice immunized with this preparation by mouth (v.o.) or intraperitoneally (i.p.) and analyzed the protection after challenge with viable parasites. BALB/c mice were immunized with three i.p. or v.o. doses of irradiated T. gondii tachyzoites. Oral challenge with ten cysts of the ME-49 or VEG strain at 90 days after the last dose resulted in high levels of protection with low parasite burden in the immunized animals. There were higher levels of specific IgG, IgA and IgM antibodies in the serum, and the i.p. immunized mice had higher levels of the high-affinity IgG and IgM antibodies than the orally immunized mice, which had more high-affinity IgA antibodies. B cells (CD19(+)), plasma cells (CD138(+)) and the CD4(+) and CD8(+) T cell populations were increased in both the blood and spleen. Cells from the spleen of the i.p. immunized mice also showed antigen-induced production of interleukin-10 (IL-10), interferon gamma (IFN-γ) and interleukin 4 (IL-4). The CD4(+) T cells, B cells and likely CD8(+) T cells from the spleens of the i.p. immunized mice proliferated with a specific antigen. The protection was correlated with the spleen and blood CD8(+) T cell, high-affinity IgG and IgM and antigen-induced IL-10 and IL-4 production. Immunization with irradiated T. gondii tachyzoites induces an immune response that is mediated by B cells and CD4(+) and CD8(+) T cells, with increased humoral and cellular immune responses that are necessary for host protection after infection. The vaccine is similar to natural infection, but free of tissue cysts; this immunity restrains infection at challenge and can be an attractive and efficient model for vaccine development in toxoplasmosis.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antibodies, Protozoan/blood
- B-Lymphocytes/immunology
- Blood/immunology
- Cell Proliferation
- Cytokines/metabolism
- Disease Models, Animal
- Immunity, Cellular
- Immunity, Humoral
- Immunoglobulin A/blood
- Immunoglobulin G/blood
- Immunoglobulin M/blood
- Injections, Intraperitoneal
- Male
- Mice, Inbred BALB C
- Protozoan Vaccines/administration & dosage
- Protozoan Vaccines/immunology
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
- Toxoplasma/immunology
- Toxoplasmosis, Animal/prevention & control
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/immunology
Collapse
Affiliation(s)
- Nahiara Esteves Zorgi
- Departamento de Parasitologia, Instituto de Ciências Biomédica, USP, Av. Prof. Lineu Prestes, 1374, Edifício Biomédicas II Cidade Universitária, São Paulo, SP, CEP: 05508-000, Brazil
- Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo, FMUSP, USP, Av. Dr. Enéas de Carvalho Aguiar, 470, 1° Andar, São Paulo, SP, CEP: 05403-000, Brazil
| | - Andrés Jimenez Galisteo
- Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo, FMUSP, USP, Av. Dr. Enéas de Carvalho Aguiar, 470, 1° Andar, São Paulo, SP, CEP: 05403-000, Brazil
| | - Maria Notomi Sato
- Departamento de Dermatologia, Instituto de Medicina Tropical de São Paulo, FMUSP, USP, Av. Dr. Enéas de Carvalho Aguiar, 470, 3° Andar, São Paulo, SP, CEP: 05403-000, Brazil
| | - Nanci do Nascimento
- Laboratório de Biologia Molecular, Instituto de Pesquisas Energéticas e Nucleares, IPEN, Rua Travessa 400, Cidade Universitária, São Paulo, SP, CEP: 05508-900, Brazil
| | - Heitor Franco de Andrade
- Departamento de Parasitologia, Instituto de Ciências Biomédica, USP, Av. Prof. Lineu Prestes, 1374, Edifício Biomédicas II Cidade Universitária, São Paulo, SP, CEP: 05508-000, Brazil.
- Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo, FMUSP, USP, Av. Dr. Enéas de Carvalho Aguiar, 470, 1° Andar, São Paulo, SP, CEP: 05403-000, Brazil.
- Department of Pathology, Faculty of Medicine, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
15
|
Prota G, Christensen D, Andersen P, Medaglini D, Ciabattini A. Peptide-specific T helper cells identified by MHC class II tetramers differentiate into several subtypes upon immunization with CAF01 adjuvanted H56 tuberculosis vaccine formulation. Vaccine 2015; 33:6823-30. [PMID: 26494626 DOI: 10.1016/j.vaccine.2015.09.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 07/17/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022]
Abstract
CD4(+) T-cell priming is an essential step in vaccination due to the key role of T helper cells in driving both effector and memory immune responses. Here we have characterized in C57BL/6 mice the T helper subtype differentiation among tetramer-specific CD4(+) T cells primed by subcutaneous immunization with the tuberculosis vaccine antigen H56 plus the adjuvant CAF01. Peptide-specific population identified by the MHC class II tetramers differentiated into several T helper subtypes upon antigen encounter, and the frequency of subpopulations differed according to their localization. Th1 (CXCR3(+)T-bet(+)), Tfh (CXCR5(+)PD-1(+)Bcl-6(+)) and RORγt(+) cells were induced in the lymph nodes draining the immunization site (dLN), while Th1 cells were the predominant subtype in the spleen. In addition, CD4(+) T cells co-expressing multiple T-cell lineage-specifying transcription factors were also detected. In the lungs, most of the tetramer-binding T cells were RORγt(+), while Tfh and Th1 cells were absent. After boosting, a higher frequency of tetramer-binding cells co-expressing the markers CD44 and CD127 was detected compared to primed cells, and cells showed a prevalent Th1 phenotype in both dLN and spleens, while Tfh cells were significantly reduced. In conclusion, these data demonstrate that parenteral immunization with H56 and CAF01 elicits a distribution of antigen-specific CD4(+) T cells in both lymphoid tissues and lungs, and gives rise to multiple T helper subtypes, that differ depending on localization and following reactivation.
Collapse
Affiliation(s)
- Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy.
| |
Collapse
|
16
|
Ciabattini A, Prota G, Christensen D, Andersen P, Pozzi G, Medaglini D. Characterization of the Antigen-Specific CD4(+) T Cell Response Induced by Prime-Boost Strategies with CAF01 and CpG Adjuvants Administered by the Intranasal and Subcutaneous Routes. Front Immunol 2015; 6:430. [PMID: 26379666 PMCID: PMC4551867 DOI: 10.3389/fimmu.2015.00430] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/09/2015] [Indexed: 11/13/2022] Open
Abstract
The design of heterologous prime-boost vaccine combinations that optimally shape the immune response is of critical importance for the development of next generation vaccines. Here, we tested different prime-boost combinations using the tuberculosis vaccine antigen H56 with CAF01 or CpG ODN 1826 adjuvants, administered by the parenteral and nasal routes. Using peptide-MHC class II tetramers, antigen-specific CD4(+) T cells were tracked following primary and booster immunizations. Both parenteral priming with H56 plus CAF01 and nasal priming with H56 plus CpG elicited significant expansion of CD4(+) tetramer-positive T cells in the spleen; however, only parenterally primed cells responded to booster immunization. Subcutaneous (SC) priming with H56 and CAF01 followed by nasal boosting with H56 and CpG showed the greater expansion of CD4(+) tetramer-positive T cells in the spleen and lungs compared to all the other homologous and heterologous prime-boost combinations. Nasal boosting exerted a recruitment of primed CD4(+) T cells into lungs that was stronger in subcutaneously than nasally primed mice, in accordance with different chemokine receptor expression induced by primary immunization. These data demonstrate that SC priming is fundamental for eliciting CD4(+) T cells that can be efficiently boosted by the nasal route and results in the recruitment of antigen-experienced cells into the lungs. Combination of different vaccine formulations and routes of delivery for priming and boosting is a strategic approach for improving and directing vaccine-induced immune responses.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| | - Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| | - Dennis Christensen
- Department for Infectious Disease Immunology, Statens Serum Institut , Copenhagen , Denmark
| | - Peter Andersen
- Department for Infectious Disease Immunology, Statens Serum Institut , Copenhagen , Denmark
| | - Gianni Pozzi
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| |
Collapse
|
17
|
Boianelli A, Pettini E, Prota G, Medaglini D, Vicino A. A Stochastic Model for CD4+ T Cell Proliferation and Dissemination Network in Primary Immune Response. PLoS One 2015; 10:e0135787. [PMID: 26301680 PMCID: PMC4547705 DOI: 10.1371/journal.pone.0135787] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/27/2015] [Indexed: 01/29/2023] Open
Abstract
The study of the initial phase of the adaptive immune response after first antigen encounter provides essential information on the magnitude and quality of the immune response. This phase is characterized by proliferation and dissemination of T cells in the lymphoid organs. Modeling and identifying the key features of this phenomenon may provide a useful tool for the analysis and prediction of the effects of immunization. This knowledge can be effectively exploited in vaccinology, where it is of interest to evaluate and compare the responses to different vaccine formulations. The objective of this paper is to construct a stochastic model based on branching process theory, for the dissemination network of antigen-specific CD4+ T cells. The devised model is validated on in vivo animal experimental data. The model presented has been applied to the vaccine immunization context making references to simple proliferation laws that take into account division, death and quiescence, but it can also be applied to any context where it is of interest to study the dynamic evolution of a population.
Collapse
Affiliation(s)
- Alessandro Boianelli
- Systems Medicine of Infectious Diseases group and Braunschweig Integrated Centre of Systems Biology, Department of Systems Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- * E-mail:
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologie, Dipartimento di Biotecnologie Mediche, Università di Siena, Viale Bracci 1, 53100 Siena, Italy
| | - Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologie, Dipartimento di Biotecnologie Mediche, Università di Siena, Viale Bracci 1, 53100 Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologie, Dipartimento di Biotecnologie Mediche, Università di Siena, Viale Bracci 1, 53100 Siena, Italy
| | - Antonio Vicino
- Dipartimento di Ingegneria dell’Informazione e Science Matematiche, Università di Siena, Via Roma 56, 53100 Siena, Italy
| |
Collapse
|
18
|
Wang Y, Sui Y, Kato S, Hogg AE, Steel JC, Morris JC, Berzofsky JA. Vaginal type-II mucosa is an inductive site for primary CD8⁺ T-cell mucosal immunity. Nat Commun 2015; 6:6100. [PMID: 25600442 PMCID: PMC4348041 DOI: 10.1038/ncomms7100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/15/2014] [Indexed: 02/07/2023] Open
Abstract
The structured lymphoid tissues are considered the only inductive sites where primary T-cell immune responses occur. The naïve T cells in structured lymphoid tissues, once being primed by antigen-bearing dendritic cells, differentiate into memory T cells and traffic back to the mucosal sites through the bloodstream. Contrary to this belief, here we show that the vaginal type-II mucosa itself, despite the lack of structured lymphoid tissues, can act as an inductive site during primary CD8(+) T-cell immune responses. We provide evidence that the vaginal mucosa supports both the local immune priming of naïve CD8(+) T cells and the local expansion of antigen-specific CD8(+) T cells, thereby demonstrating a different paradigm for primary mucosal T-cell immune induction.
Collapse
Affiliation(s)
- Yichuan Wang
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Shingo Kato
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Alison E Hogg
- 1] Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA [2] Aeras, 1405 Research Boulevard, Rockville, Maryland 20850, USA
| | - Jason C Steel
- 1] The University of Queensland, Brisbane, Queensland 4120, Australia [2] Gallipoli Medical Research Foundation, Greenslopes, Queensland 4120, Australia
| | - John C Morris
- University of Cincinnati Cancer Institute, Cincinnati, Ohio 45267, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
19
|
Allan ERO, Tailor P, Balce DR, Pirzadeh P, McKenna NT, Renaux B, Warren AL, Jirik FR, Yates RM. NADPH Oxidase Modifies Patterns of MHC Class II–Restricted Epitopic Repertoires through Redox Control of Antigen Processing. THE JOURNAL OF IMMUNOLOGY 2014; 192:4989-5001. [DOI: 10.4049/jimmunol.1302896] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
20
|
Gerlini A, Colomba L, Furi L, Braccini T, Manso AS, Pammolli A, Wang B, Vivi A, Tassini M, van Rooijen N, Pozzi G, Ricci S, Andrew PW, Koedel U, Moxon ER, Oggioni MR. The role of host and microbial factors in the pathogenesis of pneumococcal bacteraemia arising from a single bacterial cell bottleneck. PLoS Pathog 2014; 10:e1004026. [PMID: 24651834 PMCID: PMC3961388 DOI: 10.1371/journal.ppat.1004026] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 02/10/2014] [Indexed: 01/27/2023] Open
Abstract
The pathogenesis of bacteraemia after challenge with one million pneumococci of three isogenic variants was investigated. Sequential analyses of blood samples indicated that most episodes of bacteraemia were monoclonal events providing compelling evidence for a single bacterial cell bottleneck at the origin of invasive disease. With respect to host determinants, results identified novel properties of splenic macrophages and a role for neutrophils in early clearance of pneumococci. Concerning microbial factors, whole genome sequencing provided genetic evidence for the clonal origin of the bacteraemia and identified SNPs in distinct sub-units of F0/F1 ATPase in the majority of the ex vivo isolates. When compared to parental organisms of the inoculum, ex-vivo pneumococci with mutant alleles of the F0/F1 ATPase had acquired the capacity to grow at low pH at the cost of the capacity to grow at high pH. Although founded by a single cell, the genotypes of pneumococci in septicaemic mice indicate strong selective pressure for fitness, emphasising the within-host complexity of the pathogenesis of invasive disease.
Collapse
Affiliation(s)
- Alice Gerlini
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
| | - Leonarda Colomba
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
| | - Leonardo Furi
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
| | - Tiziana Braccini
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
| | - Ana Sousa Manso
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
| | - Andrea Pammolli
- Department of Pathophysiology, Experimental Medicine and Public Health, University of Siena, Siena, Italy
| | - Bo Wang
- Department of Mathematics, University of Leicester, Leicester, United Kingdom
| | | | | | - Nico van Rooijen
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Gianni Pozzi
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
- UOC Batteriologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Susanna Ricci
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
| | - Peter W. Andrew
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Uwe Koedel
- Department of Neurology, Ludwig-Maximilians University of Munich, München, Germany
| | - E. Richard Moxon
- Division of Medical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Marco R. Oggioni
- LAMMB, Department of Biotechnology, University of Siena, Siena, Italy
- UOC Batteriologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
21
|
Pettini E, Prota G, Ciabattini A, Boianelli A, Fiorino F, Pozzi G, Vicino A, Medaglini D. Vaginal immunization to elicit primary T-cell activation and dissemination. PLoS One 2013; 8:e80545. [PMID: 24349003 PMCID: PMC3857820 DOI: 10.1371/journal.pone.0080545] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/14/2013] [Indexed: 01/30/2023] Open
Abstract
Primary T-cell activation at mucosal sites is of utmost importance for the development of vaccination strategies. T-cell priming after vaginal immunization, with ovalbumin and CpG oligodeoxynucleotide adjuvant as model vaccine formulation, was studied in vivo in hormone-synchronized mice and compared to the one induced by the nasal route. Twenty-four hours after both vaginal or nasal immunization, antigen-loaded dendritic cells were detected within the respective draining lymph nodes. Vaginal immunization elicited a strong recruitment of antigen-specific CD4(+) T cells into draining lymph nodes that was more rapid than the one observed following nasal immunization. T-cell clonal expansion was first detected in iliac lymph nodes, draining the genital tract, and proliferated T cells disseminated towards distal lymph nodes and spleen similarly to what observed following nasal immunization. T cells were indeed activated by the antigen encounter and acquired homing molecules essential to disseminate towards distal lymphoid organs as confirmed by the modulation of CD45RB, CD69, CD44 and CD62L marker expression. A multi-type Galton Watson branching process, previously used for in vitro analysis of T-cell proliferation, was applied to model in vivo CFSE proliferation data in draining lymph nodes 57 hours following immunization, in order to calculate the probabilistic decision of a cell to enter in division, rest in quiescence or migrate/die. The modelling analysis indicated that the probability of a cell to proliferate was higher following vaginal than nasal immunization. All together these data show that vaginal immunization, despite the absence of an organized mucosal associated inductive site in the genital tract, is very efficient in priming antigen-specific CD4(+) T cells and inducing their dissemination from draining lymph nodes towards distal lymphoid organs.
Collapse
Affiliation(s)
- Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Alessandro Boianelli
- Dipartimento di Ingegneria dell'Informazione (DII), Centro per lo Studio dei Sistemi Complessi (CSC), Università di Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Gianni Pozzi
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Antonio Vicino
- Dipartimento di Ingegneria dell'Informazione (DII), Centro per lo Studio dei Sistemi Complessi (CSC), Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
- * E-mail:
| |
Collapse
|
22
|
Ciabattini A, Pettini E, Medaglini D. CD4(+) T Cell Priming as Biomarker to Study Immune Response to Preventive Vaccines. Front Immunol 2013; 4:421. [PMID: 24363656 PMCID: PMC3850413 DOI: 10.3389/fimmu.2013.00421] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/20/2013] [Indexed: 01/10/2023] Open
Abstract
T cell priming is a critical event in the initiation of the immune response to vaccination since it deeply influences both the magnitude and the quality of the immune response induced. CD4(+) T cell priming, required for the induction of high-affinity antibodies and immune memory, represents a key target for improving and modulating vaccine immunogenicity. A major challenge in the study of in vivo T cell priming is due to the low frequency of antigen-specific T cells. This review discusses the current knowledge on antigen-specific CD4(+) T cell priming in the context of vaccination, as well as the most advanced tools for the characterization of the in vivo T cell priming and the opportunities offered by the application of systems biology.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| |
Collapse
|
23
|
Segawa T, Saeki A, Hasebe A, Arimoto T, Kataoka H, Yokoyama A, Kawanami M, Shibata KI. Differences in recognition of wild-type and lipoprotein-deficient strains of oralStreptococci in vitroandin vivo. Pathog Dis 2013; 68:65-77. [DOI: 10.1111/2049-632x.12049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 01/11/2023] Open
Affiliation(s)
| | - Ayumi Saeki
- Division of Oral Molecular Microbiology; Department of Oral Pathobiological Science; Hokkaido University Graduate School of Dental Medicine; Kita-ku; Sapporo; Japan
| | - Akira Hasebe
- Division of Oral Molecular Microbiology; Department of Oral Pathobiological Science; Hokkaido University Graduate School of Dental Medicine; Kita-ku; Sapporo; Japan
| | - Takafumi Arimoto
- Department of Oral Microbiology; Showa University School of Dentistry; Shinagawa-ku; Tokyo; Japan
| | - Hideo Kataoka
- Department of Oral Microbiology; Showa University School of Dentistry; Shinagawa-ku; Tokyo; Japan
| | - Atsuro Yokoyama
- Division of Oral Functional Science; Department of Oral Functional Prosthodontics; Hokkaido University Graduate School of Dental Medicine; Kita-ku; Sapporo; Japan
| | - Masamitsu Kawanami
- Division of Oral Health Science; Department of Periodontology and Endodontology; Hokkaido University Graduate School of Dental Medicine; Kita-ku; Sapporo; Japan
| | - Ken-ichiro Shibata
- Division of Oral Molecular Microbiology; Department of Oral Pathobiological Science; Hokkaido University Graduate School of Dental Medicine; Kita-ku; Sapporo; Japan
| |
Collapse
|
24
|
Fiorino F, Pettini E, Pozzi G, Medaglini D, Ciabattini A. Prime-boost strategies in mucosal immunization affect local IgA production and the type of th response. Front Immunol 2013; 4:128. [PMID: 23755051 PMCID: PMC3665932 DOI: 10.3389/fimmu.2013.00128] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
Combinations of different delivery routes for priming and boosting represent vaccination strategies that can modulate magnitude, quality, and localization of the immune response. A murine model was used to study T cell clonal expansion following intranasal (IN) or subcutaneous (SC) priming, and secondary immune responses after boosting by either homologous or heterologous routes. T cell primary activation was studied by using the adoptive transfer model of ovalbumin-specific transgenic CD4(+) T cells. Both IN and SC immunization efficiently elicited, in the respective draining lymph nodes, primary clonal expansion of antigen-specific CD4(+) T cells that disseminated toward distal lymph nodes (mesenteric and iliac) and the spleen. After boosting, a significant serum IgG response was induced in all groups independent of the combination of immunization routes used, while significant levels of local IgA were detected only in mice boosted by the IN route. Mucosal priming drove a stronger Th1 polarization than the systemic route, as shown by serum IgG subclass analysis. IFN-gamma production was observed in splenocytes of all groups, while prime-boost vaccine combinations that included the mucosal route, yielded higher levels of IL-17. Memory lymphocytes were identified in both spleen and draining lymph nodes in all immunized mice, with the highest number of IL-2 producing cells detected in mice primed and boosted by the nasal route. This work shows the critical role of immunization routes in modulating quality and localization of immune responses in prime-boost vaccine strategies.
Collapse
Affiliation(s)
- Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia, Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| | | | | | | | | |
Collapse
|
25
|
Deng YH, He HY, Zhang BS. Evaluation of protective efficacy conferred by a recombinant Mycobacterium bovis BCG expressing a fusion protein of Ag85A-ESAT-6. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2013; 47:48-56. [PMID: 23357605 DOI: 10.1016/j.jmii.2012.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 11/01/2012] [Accepted: 11/27/2012] [Indexed: 11/26/2022]
Abstract
BACKGROUND We previously constructed a recombinant bacille Calmette-Guérin (rBCG-AE) strain that could express a fused Ag85A-ESAT-6 protein. That study suggested that the rBCG-AE strain was able to induce a higher titer of antibody and elicit a more long-lived and stronger Th1-type cellular immune responses than the parental BCG strain, the rBCG-A strain (i.e., expressing Ag85A), or the rBCG-E strain (i.e., expressing ESAT-6). METHODS In the current study, we further investigated the strain's protective efficacy against Mycobacterium tuberculosis H37Rv infection in BALB/c mice through evaluating organ bacterial loads, lung histopathology, lung immunohistochemistry, and net weight gain or loss by using conventional BCG, rBCG-A, and rBCG-E as the controls. RESULTS From the 3rd to 9th weeks after the challenge infection, the bacterial counts were significantly lower in tissues (e.g., spleen and lung tissues) in the mice immunized with rBCG-AE than in the control group, but were higher than the counts in the BCG group. The pathological damage in the lung tissues of the rBCG-AE group gradually improved from the 6th to 9th weeks after being infected with M. tuberculosis H37Rv, but the score of pathological changes in the rBCG-AE group was obviously higher than the score in the BCG group. There was no difference in the percentage of IFN-γ and iNOS positive cells in the lung tissues of the rBCG-AE and BCG groups. CONCLUSION The results suggest that rBCG-AE can not promote protective efficacy against M. tuberculosis H37Rv infection, compared to the BCG vaccine.
Collapse
Affiliation(s)
- Yi-Hao Deng
- Department of Human Anatomy, College of Preclinical Medicine, Dali University, Dali 671000, China.
| | - Hong-Yun He
- Department of Human Anatomy, College of Preclinical Medicine, Dali University, Dali 671000, China
| | - Ben-Si Zhang
- Department of Human Anatomy, College of Preclinical Medicine, Dali University, Dali 671000, China
| |
Collapse
|
26
|
Immunization with the conjugate vaccine Vi-CRM₁₉₇ against Salmonella typhi induces Vi-specific mucosal and systemic immune responses in mice. Vaccine 2012; 30:6111-4. [PMID: 22705173 DOI: 10.1016/j.vaccine.2012.05.081] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 05/19/2012] [Accepted: 05/30/2012] [Indexed: 11/21/2022]
Abstract
Typhoid fever is a public health problem, especially among young children in developing countries. To address this need, a glycoconjugate vaccine Vi-CRM₁₉₇, composed of the polysaccharide antigen Vi covalently conjugated to the non-toxic mutant of diphtheria toxin CRM₁₉₇, is under development. Here, we assessed the antibody and cellular responses, both local and systemic, following subcutaneous injection of Vi-CRM₁₉₇. The glycoconjugate elicited Vi-specific serum IgG titers significantly higher than unconjugated Vi, with prevalence of IgG1 that persisted for at least 60 days after immunization. Vi-specific IgG, but not IgA, were present in intestinal washes. Lymphocytes proliferation after restimulation with Vi-CRM₁₉₇ was observed in spleen and mesenteric lymph nodes. These data confirm the immunogenicity of Vi-CRM₁₉₇ and demonstrate that the vaccine-specific antibody and cellular immune responses are present also in the intestinal tract, thus strengthening the suitability of Vi-CRM₁₉₇ as a promising candidate vaccine against Salmonella Typhi.
Collapse
|
27
|
Deng Y, Bao L, Yang X. Evaluation of immunogenicity and protective efficacy against Mycobacterium tuberculosis infection elicited by recombinant Mycobacterium bovis BCG expressing human Interleukin-12p70 and Early Secretory Antigen Target-6 fusion protein. Microbiol Immunol 2012; 55:798-808. [PMID: 21831202 DOI: 10.1111/j.1348-0421.2011.00376.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
ESAT-6 protein of Mycobacterium tuberculosis is absent in Mycobacterium bovis BCG and Mycobacterium microti and has been demonstrated to stimulate strong cell-mediated immunity. IL-12 can play crucial roles in regulating IFN-γ production and Th1 effectors production. In this study, we constructed three rBCG vaccines that could express proteins of human IL-12p70 and/or ESAT-6 and evaluated their immunogenicity and protective efficacy in mice. Our experiments illustrated that the rBCG-IE (expressing a fusion protein of human IL-12p70 and ESAT-6) was capable of inducing stronger Th1 type cell-mediated immune responses than conventional BCG, or rBCG-I (expressing human IL-12p70), or rBCG-E (expressing ESAT-6). However, the results of protective experiments showed that rBCG-IE could only confer similar and even lower protective efficacy against M. tuberculosis H37Rv infection compared with BCG vaccine.
Collapse
Affiliation(s)
- Yihao Deng
- Laboratory of Infection and Immunity, West China Center of Medical Ssciences, Sichuan University, Chengdu 610041, China
| | | | | |
Collapse
|
28
|
Sable SB, Cheruvu M, Nandakumar S, Sharma S, Bandyopadhyay K, Kellar KL, Posey JE, Plikaytis BB, Amara RR, Shinnick TM. Cellular immune responses to nine Mycobacterium tuberculosis vaccine candidates following intranasal vaccination. PLoS One 2011; 6:e22718. [PMID: 21799939 PMCID: PMC3143185 DOI: 10.1371/journal.pone.0022718] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 06/29/2011] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The identification of Mycobacterium tuberculosis vaccines that elicit a protective immune response in the lungs is important for the development of an effective vaccine against tuberculosis. METHODS AND PRINCIPAL FINDINGS In this study, a comparison of intranasal (i.n.) and subcutaneous (s.c.) vaccination with the BCG vaccine demonstrated that a single moderate dose delivered intranasally induced a stronger and sustained M. tuberculosis-specific T-cell response in lung parenchyma and cervical lymph nodes of BALB/c mice than vaccine delivered subcutaneously. Both BCG and a multicomponent subunit vaccine composed of nine M. tuberculosis recombinant proteins induced strong antigen-specific T-cell responses in various local and peripheral immune compartments. Among the nine recombinant proteins evaluated, the alanine proline rich antigen (Apa, Rv1860) was highly antigenic following i.n. BCG and immunogenic after vaccination with a combination of the nine recombinant antigens. The Apa-induced responses included induction of both type 1 and type 2 cytokines in the lungs as evaluated by ELISPOT and a multiplexed microsphere-based cytokine immunoassay. Of importance, i.n. subunit vaccination with Apa imparted significant protection in the lungs and spleen of mice against M. tuberculosis challenge. Despite observed differences in the frequencies and location of specific cytokine secreting T cells both BCG vaccination routes afforded comparable levels of protection in our study. CONCLUSION AND SIGNIFICANCE Overall, our findings support consideration and further evaluation of an intranasally targeted Apa-based vaccine to prevent tuberculosis.
Collapse
Affiliation(s)
- Suraj B Sable
- Division of TB Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ciabattini A, Pettini E, Fiorino F, Prota G, Pozzi G, Medaglini D. Distribution of primed T cells and antigen-loaded antigen presenting cells following intranasal immunization in mice. PLoS One 2011; 6:e19346. [PMID: 21559409 PMCID: PMC3084830 DOI: 10.1371/journal.pone.0019346] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 03/28/2011] [Indexed: 10/31/2022] Open
Abstract
Priming of T cells is a key event in vaccination, since it bears a decisive influence on the type and magnitude of the immune response. T-cell priming after mucosal immunization via the nasal route was studied by investigating the distribution of antigen-loaded antigen presenting cells (APCs) and primed antigen-specific T cells. Nasal immunization studies were conducted using the model protein antigen ovalbumin (OVA) plus CpG oligodeoxynucleotide adjuvant. Trafficking of antigen-specific primed T cells was analyzed in vivo after adoptive transfer of OVA-specific transgenic T cells in the presence or absence of fingolimod, a drug that causes lymphocytes sequestration within lymph nodes. Antigen-loaded APCs were observed in mediastinal lymph nodes, draining the respiratory tract, but not in distal lymph nodes. Antigen-specific proliferating T cells were first observed within draining lymph nodes, and later in distal iliac and mesenteric lymph nodes and in the spleen. The presence at distal sites was due to migration of locally primed T cells as shown by fingolimod treatment that caused a drastic reduction of proliferated T cells in non-draining lymph nodes and an accumulation of extensively divided T cells within draining lymph nodes. Homing of nasally primed T cells in distal iliac lymph nodes was CD62L-dependent, while entry into mesenteric lymph nodes depended on both CD62L and α4β7, as shown by in vivo antibody-mediated inhibition of T-cell trafficking. These data, elucidating the trafficking of antigen-specific primed T cells to non-draining peripheral and mucosa-associated lymph nodes following nasal immunization, provide relevant insights for the design of vaccination strategies based on mucosal priming.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie, Università di Siena, Siena, Italy
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie, Università di Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie, Università di Siena, Siena, Italy
| | - Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie, Università di Siena, Siena, Italy
| | - Gianni Pozzi
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie, Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie, Università di Siena, Siena, Italy
- * E-mail:
| |
Collapse
|
30
|
Lian ZR, Xu YF, Wang XB, Gong JP, Liu ZJ. Suppression of histone deacetylase 11 promotes expression of IL-10 in Kupffer cells and induces tolerance following orthotopic liver transplantation in rats. J Surg Res 2011; 174:359-68. [PMID: 21392795 DOI: 10.1016/j.jss.2010.12.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 12/13/2010] [Accepted: 12/22/2010] [Indexed: 11/17/2022]
Abstract
BACKGROUND Suppression of histone deacetylase 11 (HDAC11) can promote IL-10 expression in mouse macrophages RAW264.7 and induce immune tolerance. This study is to further investigate the role of HDAC11 in tolerance induction via Kupffer cells (KCs) following orthotopic liver transplantation (OLT) in rats. MATERIALS AND METHODS KCs isolated from BALB/c mice were divided into pHDAC11, adHDAC11, and pCV group (treated with HADC11-shRNA, adenovirus encoding HDAC11, and control vector, respectively). IL-10 expression was determined after lipopolysaccharide treatment. The expression of MHC-II and co-stimulatory molecules on KCs surface was evaluated by flow cytometry. T cell proliferation was measured by [(3)H]-thymidine incorporation after culturing with aforementioned three groups, treated KCs, respectively. OLT was performed in rats after Ad-HDAC11 and pHDAC11 treatment. Blood samples were collected for biochemical studies, and postoperative survival was examined. RESULTS IL-10 expression was inhibited and promoted by Ad-HDAC11 and HDAC11-shRNA in KCs, respectively. MHC-II and co-stimulatory molecules on KCs surface as well as T cell proliferation were significantly inhibited and induced in pHDAC11 and Ad-HDAC11 compared with pCV, respectively. Serum IL-2, TNF-α, and IFN-γ levels were significantly lower in pHDAC11 and higher in Ad-HDAC11 compared with pCV, respectively, while IL-4 and IL-10 were the reverse. Postoperative survival, liver function, and histology were different among the three groups. CONCLUSIONS Suppression of HDAC11 can promote IL-10 expression in KCs and induce tolerance following OLT in rats. Consequently, HDAC11 may be a key component of this immune regulation system and a promising target for development of novel drugs of gene therapy for inducing tolerance in clinical liver transplantation.
Collapse
Affiliation(s)
- Zheng-rong Lian
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
31
|
Vaccine adjuvants: A priority for vaccine research. Vaccine 2010; 28:2363-6. [DOI: 10.1016/j.vaccine.2009.12.084] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2009] [Accepted: 12/25/2009] [Indexed: 01/03/2023]
|
32
|
Ciabattini A, Pettini E, Arsenijevic S, Pozzi G, Medaglini D. Intranasal immunization with vaccine vector Streptococcus gordonii elicits primed CD4+ and CD8+ T cells in the genital and intestinal tracts. Vaccine 2009; 28:1226-33. [PMID: 19945415 DOI: 10.1016/j.vaccine.2009.11.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 10/30/2009] [Accepted: 11/09/2009] [Indexed: 01/01/2023]
Abstract
Generation of primed T cells is crucial for the development of optimal vaccination strategies. Using a TCR-transgenic CD4(+) and CD8(+) T cell adoptive transfer model, we demonstrate that a single nasal immunization with recombinant Streptococcus gordonii induces antigen-specific primed T cells in lymph nodes draining the genital and intestinal tracts with about 80% of CD4(+) and 50% of CD8(+) proliferating cells. T cell clonal expansion was also observed in cervical lymph nodes, draining the immunization site, and in the spleen. The modulation of CD44 and CD45RB marker expression indicated that proliferating T cells were activated. Proliferation in distal mesenteric and iliac lymph nodes and in the spleen was observed 5 days after nasal immunization, while in draining cervical lymph nodes proliferation peaked already at day 3. The division profile of transgenic T cells observed in iliac and mesenteric lymph nodes was discontinuous, showing the lack of early cell divisions. The kinetics of T cell clonal expansion, the discontinuous division profile and the modulation of migration markers such as CD62L suggest that activated antigen-specific T cells disseminate from the immunization site to distal intestinal and genital tracts. These data demonstrate the efficacy of nasal immunization with recombinant S. gordonii in eliciting CD4(+) and CD8(+) T cell priming not only in draining sites, but also in the genital and intestinal tracts and in the spleen.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biologia Molecolare, Università di Siena, 53100 Siena, Italy
| | | | | | | | | |
Collapse
|
33
|
A novel DNA vaccine containing multiple TB-specific epitopes casted in a natural structure (ECANS) confers protective immunity against pulmonary mycobacterial challenge. Vaccine 2009; 27:5313-9. [PMID: 19615961 PMCID: PMC7115364 DOI: 10.1016/j.vaccine.2009.06.093] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 06/09/2009] [Accepted: 06/29/2009] [Indexed: 11/21/2022]
Abstract
Epitope-based DNA vaccines designed to induce T cell responses specific for Mycobacterium tuberculosis (M. tb) are being developed as a means of addressing vaccine potency. In this study, we predicted 4 T cell epitopes from ESAT-6, Ag85A/B and CFP-10 antigens and constructed an ECANS (epitopes casted in a natural structure) DNA vaccine by inserting the epitope DNA segments separately into the gene backbone of M. tb-derived HSP65 (heat shock protein 65) carrier. The immunogenicity and protective efficacy of pECANS DNA vaccine were assessed in BALB/c mice after intramuscular immunization with 4 doses of 50 microg ECANS DNA and followed by mycobaterial challenge 4 weeks after the last immunization. Compared to plasmid encoding HSP65, pECANS DNA immunization elicited remarkably higher levels of IFN-gamma production by both CD4(+) and CD8(+) T cells, which were coupled with higher frequencies of antigen-specific T cells and higher CTL activity. Significantly enhanced levels of Th1 cytokines (IFN-gamma and IL-12) and increased serum IgG2a/IgG1 ratio were also noted, indicating a predominant Th1 immune response achieved by pECANS DNA immunization. In the consequence, a better protection against Mycobacterium bovis BCG challenge was achieved which was evidenced by reduced bacterial loads in lungs and spleens and profound attenuation of lung inflammation and injury. Our results suggested that multi-T cell-epitope based ECANS gene vaccine induced T cell response to multiple T cell epitopes and led to enhanced protection against mycobacterial challenge. This strategy might be a useful platform to design multi-T cell epitope-based vaccine against M. tb infection.
Collapse
|
34
|
Vendetti S, Riccomi A, Negri DRM, Veglia F, Sciaraffia E, De Magistris MT. Development of antigen-specific T cells in mediastinal lymph nodes after intranasal immunization. Methods 2009; 49:334-9. [PMID: 19426804 DOI: 10.1016/j.ymeth.2009.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 04/16/2009] [Accepted: 04/23/2009] [Indexed: 10/20/2022] Open
Abstract
The power of cholera toxin (CT) as an effective mucosal adjuvant is well established. Because of the high toxicity of CT, its clinical use is unlikely. Therefore, the need to identify effective and non toxic mucosal adjuvants for human use is important. For this purpose, CT is largely used as a reference molecule for testing the efficacy of new candidate adjuvants in animal models. Here, we evaluated the kinetics and the localization of antigen-specific humoral and cellular immune responses elicited by intranasal immunization with tetanus toxoid antigen in the presence of CT. We show that an antigen-specific cellular immune response localized in the mediastinal lymph nodes can be observed already 1 week after the first immunization. The induction of an appreciable titer of an antibody-specific immune response was assessed after two immunizations. Therefore, we suggest that the efficacy of new candidate mucosal adjuvants can be tested by evaluating the cellular immune response in the mediastinal lymph nodes at early stages of immunization.
Collapse
Affiliation(s)
- Silvia Vendetti
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
35
|
Pettini E, Ciabattini A, Pozzi G, Medaglini D. Adoptive transfer of transgenic T cells to study mucosal adjuvants. Methods 2009; 49:340-5. [PMID: 19409994 DOI: 10.1016/j.ymeth.2009.03.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 02/24/2009] [Accepted: 03/09/2009] [Indexed: 10/20/2022] Open
Abstract
The study of the initiation and regulation of T-cell responses to vaccine antigens is of primary importance in the rational design of mucosal adjuvants. The detection in vivo of T-cell priming following immunization can be performed by using the adoptive transfer model of naïve antigen-specific transgenic T cells into immunocompetent mice. In this work, we discuss the applications of this system for detecting in vivo the primary antigen-specific clonal expansion, the phenotype, and the effector function of transgenic T cells following mucosal immunization. OVA and the mucosal adjuvant CTB were used as a model vaccine formulation and administered by the nasal route to study T-cell priming. T helper and T cytotoxic primary proliferation and expression of activation and migration markers was observed both in draining and distal sites. This method proved to be a powerful tool to study the efficacy of mucosal adjuvants in enhancing T-cell priming.
Collapse
Affiliation(s)
- Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia, Dipartimento di Biologia Molecolare, Università di Siena, 53100 Siena, Italy
| | | | | | | |
Collapse
|
36
|
The European effort towards the development of mucosal vaccines for poverty-related diseases. Vaccine 2009; 27:2641-8. [DOI: 10.1016/j.vaccine.2009.02.070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Revised: 02/10/2009] [Accepted: 02/18/2009] [Indexed: 12/20/2022]
|