1
|
Jia Z, Zhou X, Liu J, De X, Li Y, Yang Z, Duan H, Wang F, Ge J. Immune enhancement of rhamnolipid/manganese calcium phosphate mineralized nanoparticle: A promising subunit antigen delivery system. Int J Biol Macromol 2024; 282:137239. [PMID: 39491710 DOI: 10.1016/j.ijbiomac.2024.137239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
The use of biomimetic mineralization strategy is promising to solve the problem of poor stability and immune effect of subunit antigens. However, non-specifically inducing protein mineralization is still a challenge. we hypothesized that rhamnolipids with both protein and metal binding capacity could be used to develop more functional and biocompatible calcium mineralized nanoparticle (RMCP). The results show that rhamnolipids synergistically enhanced the mineralization of protein with manganese ions and improved 21 % the loading antigens of RMCP compared to manganese calcium phosphate nanoparticles. Transmission electron microscopy (TEM) and Dynamic Light Scattering (DLS) showed particle size of RMCP is 260 ± 12.1 nm with spherical morphology. In vitro experiments have shown that RMCP effectively activate immune cells through the cGAS-STING and NLRP3 pathways and demonstrated a higher level of cytokines in RAW264.7 Macrophages. In vivo, RMCP triggered an increased IgG titer with 16.5-fold IgG2a/IgG1 ratio compared to the aluminum adjuvant which improved the recovery status after challenge in mice. We used biological surfactants for the first time to enhance the biomimetic mineralization process of subunit antigen, which provides a new approach for constructing calcium-based biocompatible antigen delivery vectors, helping to develop a new generation of stable, efficient, and safe subunit vaccines.
Collapse
Affiliation(s)
- Zheng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Xinyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Jingjing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Xinqi De
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Yifan Li
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Zaixing Yang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Haoyuan Duan
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Fang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China.
| |
Collapse
|
2
|
Saki M, De Villiers H, Ntsapi C, Tiloke C. The Hepatoprotective Effects of Moringa oleifera against Antiretroviral-Induced Cytotoxicity in HepG 2 Cells: A Review. PLANTS (BASEL, SWITZERLAND) 2023; 12:3235. [PMID: 37765399 PMCID: PMC10537654 DOI: 10.3390/plants12183235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/20/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
The untreated human immunodeficiency virus (HIV), a lentivirus species that attacks immune cells (CD4+ T cells), causes acquired immunodeficiency syndrome (AIDS). HIV-positive people manage HIV/AIDS by using antiretroviral therapy (ART). The ART treatment regimen contains two nucleoside reverse transcriptase inhibitors (NRTIs) and one non-nucleoside reverse transcriptase inhibitor/integrase strand transfer inhibitor. Tenofovir, an NRTI approved for managing HIV infection, is associated with hepatic steatosis and lactic acidosis, which are linked to mitochondrial toxicity and oxidative stress. Due to side-effects associated with ART, people living with HIV often use medicinal plants or a combination of medicinal plants with ART to promote adherence and diminish the side-effects and cytotoxicity. The Moringa oleifera (MO) tree from the family of Moringaceae is among the medicinal trees studied in managing HIV/AIDS in sub-Saharan Africa. The MO tree extracts have been reported to have inhibitory activity primarily against HIV due to their bioactive compounds. However, there is a scarcity of knowledge about the use of the MO tree amongst HIV/AIDS patients receiving ART in South Africa and its effect on patient compliance and outcomes. Thus, this review aims to outline the impact of MO aqueous leaf extract on oxidative stress and antioxidant responses in human HepG2 liver cells after exposure to antiretrovirals such as tenofovir. The review will contribute to a comprehensive understanding of the potential protective effect of MO aqueous leaf extract on tenofovir-induced cytotoxicity.
Collapse
Affiliation(s)
| | | | | | - Charlette Tiloke
- Department of Basic Medical Sciences, School of Biomedical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (M.S.); (H.D.V.); (C.N.)
| |
Collapse
|
3
|
Gao Y, Gong X, Yu S, Jin Z, Ruan Q, Zhang C, Zhao K. Immune enhancement of N-2-Hydroxypropyl trimethyl ammonium chloride chitosan/carboxymethyl chitosan nanoparticles vaccine. Int J Biol Macromol 2022; 220:183-192. [PMID: 35981671 DOI: 10.1016/j.ijbiomac.2022.08.073] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 11/05/2022]
Abstract
The immunogenicity and toxicity of N-2-Hydroxypropyl trimethyl ammonium chloride chitosan/N, O-carboxymethyl chitosan nanoparticles (N-2-HACC/CMCS NPs) as a universal vaccine adjuvant/delivery system remains unclear. The present study indicated that the positively charged N-2-HACC/CMCS NPs showed a regular spherical morphology, with a particle size of 219 ± 13.72 nm, zeta potential of 37.28 ± 4.58 mV, had hemocompatibility and biodegradation. Acute toxicity, repeated dose toxicity, abnormal toxicity, muscle stimulation, whole body allergic reaction evaluation in vitro, and cytotoxicity in vivo confirmed N-2-HACC/CMCS NPs is safe and non-toxic. N-2-HACC/OVA/CMCS NPs were prepared to evaluate the immunogenicity, which showed a particle size of 248.1 ± 15.53 nm, zeta potential of 17.24 ± 1.28 mV, encapsulation efficiency of 92.43 ± 0.96 %, and loading capacity of 42.97 ± 0.07 %. Oral or intramuscular route with the N-2-HACC/OVA/CMCS NPs in mice not only induced higher IgG, IgG1, IgG2a, and sIgA antibody titers, but also significantly produced higher levels of IL-6, IL-4, IFN-γ, and TNF-α, demonstrating that the N-2-HACC/OVA/CMCS NPs enhance humoral, cellular, and mucosal immune responses. Our results not only support the N-2-HACC/CMCS NPs to be a safe and potential universal nano adjuvant/delivery system in vaccine development, especially mucosal vaccines, but also rich the database knowledge of adjuvant/delivery systems, and provide new direction to introduce more licensed adjuvants.
Collapse
Affiliation(s)
- Yuan Gao
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Xiaochen Gong
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Shuang Yu
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, Heilongjiang 150080, China
| | - Zheng Jin
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Qicheng Ruan
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Chunjing Zhang
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China.
| | - Kai Zhao
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, China; Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, Heilongjiang 150080, China; Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
4
|
Xu T, Liu L, Shi C, Liu W, Wang M, Tian L, Zheng Y, Wang H, Zheng W, He H, Xia X, Zheng X. A recombinant rabies virus expressing Echinococcus granulosus EG95 induces protective immunity in mice. Transbound Emerg Dis 2022; 69:e254-e266. [PMID: 34403194 DOI: 10.1111/tbed.14292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 11/27/2022]
Abstract
Cystic echinococcosis (CE), caused by Echinococcus granulosus (E, is a zoonosis with a worldwide distribution, resulting in heavy impact to public health and social economics. In this study, we generated a recombinant rabies virus (RABV) expressing EG95 protein of E. granulosus (LBNSE-EG95) as a bivalent candidate vaccine for use in sheep and cattle against CE and rabies, which is another severe health threat in CE-endemic areas. It was found that EG95 was successfully expressed without altering the pathogenicity of parent LBNSE vector. Further study showed that LBNSE-EG95 immunization in mice elicited activation of dendric cells (DCs) and B cells and induced Th1-/Th2-mediated cellular immune responses, leading to robust production of RABV neutralizing antibodies and high level of EG95-sepecific antibodies with more than 90% protection against CE. In addition, single dose of LBNSE-EG95 conferred full protection against lethal RABV challenge in mice. Collectively, these results suggest that the recombinant LBNSE-EG95 has the potential to be developed as an efficient bivalent vaccine for sheep and cattle use in endemic areas of CE and rabies.
Collapse
Affiliation(s)
- Tong Xu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lele Liu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chenjuan Shi
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenkai Liu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Wang
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li Tian
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ye Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongmei Wang
- Department of Biological Sciences, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Wenwen Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongbin He
- Department of Biological Sciences, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Xianzhu Xia
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xuexing Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
5
|
Zhao K, Sun B, Shi C, Sun Y, Jin Z, Hu G. Intranasal immunization with O-2'-Hydroxypropyl trimethyl ammonium chloride chitosan nanoparticles loaded with Newcastle disease virus DNA vaccine enhances mucosal immune response in chickens. J Nanobiotechnology 2021; 19:240. [PMID: 34380522 PMCID: PMC8359106 DOI: 10.1186/s12951-021-00983-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/01/2021] [Indexed: 11/16/2022] Open
Abstract
Background There has been a great interest in developing strategies for enhancing antigen delivery to the mucosal immune system as well as identifying mucosal active immunostimulating agents. To elevate the potential of O-2ʹ-Hydroxypropyl trimethyl ammonium chloride chitosan (O-2ʹ-HACC) as an adjuvant and mucosal immune delivery carrier for DNA vaccine, we prepared the O-2ʹ-HACC loaded with Newcastle disease virus (NDV) F gene plasmid DNA and C3d6 molecular adjuvant (O-2ʹ-HACC/pFDNA microparticles). Results The O-2ʹ-HACC/pFDNA exhibited a regular spherical morphology with a particle size of 202.3 ± 0.52 nm, a zeta potential of 50.8 ± 8.21 mV, encapsulation efficiency of 90.74 ± 1.10%, and a loading capacity of 49.84 ± 1.20%. The plasmid DNA could be sustainably released from the O-2ʹ-HACC/pFDNA after an initial burst release. Intranasal vaccination of chickens immunized with O-2ʹ-HACC/pFDNA not only induced higher anti-NDV IgG and sIgA antibody titers but also significantly promoted lymphocyte proliferation and produced higher levels of IL-2, IL-4, IFN-γ, CD4+, and CD8 + T lymphocytes compared with the NDV commercial live attenuated vaccine. Intranasal delivery of the O-2ʹ-HACC/pFDNA enhanced humoral, cellular, and mucosal immune responses and protected chickens from the infection of highly virulent NDV compared with the intramuscular delivery. Conclusions Collectively, our findings indicated that the O-2ʹ-HACC could be used as a vaccine adjuvant and delivery system for mucosal immunity and have an immense application promise. Graphic Abstract ![]()
Collapse
Affiliation(s)
- Kai Zhao
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, 318000, China. .,Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China.
| | - Beini Sun
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China
| | - Ci Shi
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China
| | - Yanwei Sun
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin, 150080, China
| | - Zheng Jin
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, 318000, China.,Key Laboratory of Chemical Engineering Process and Technology for High-Efficiency Conversion, College of Chemistry and Material Sciences, Heilongjiang University, Harbin, 150080, China
| | - Gaowei Hu
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, 318000, China
| |
Collapse
|
6
|
Moore TC, Hasenkrug KJ. B-Cell Control of Regulatory T Cells in Friend Virus Infection. J Mol Biol 2021; 433:166583. [PMID: 32598936 DOI: 10.1016/j.jmb.2020.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022]
Abstract
B lymphocytes have well-established effector roles during viral infections, including production of antibodies and functioning as antigen-presenting cells for CD4+ and CD8+ T cells. B cells have also been shown to regulate immune responses and induce regulatory T cells (Tregs). In the Friend virus (FV) model, Tregs are known to inhibit effector CD8+ T-cell responses and contribute to virus persistence. Recent work has uncovered a role for B cells in the induction and activation of Tregs during FV infection. In addition to inducing Tregs, B cell antibody production and antigen-presenting cell activity is a target of Treg suppression. This review focuses on the dynamic interactions between B cells and Tregs during FV infection.
Collapse
Affiliation(s)
- Tyler C Moore
- College of Science and Technology, Bellevue University, 1000 Galvin Road South, Bellevue, NE 68005, USA.
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 S. 4th Street, Hamilton, MT 59840, USA.
| |
Collapse
|
7
|
Zhang W, Cheng N, Wang Y, Zheng X, Zhao Y, Wang H, Wang C, Han Q, Gao Y, Shan J, Yang S, Xia X. Adjuvant activity of PCP-II, a polysaccharide from Poria cocos, on a whole killed rabies vaccine. Virus Res 2019; 270:197638. [PMID: 31173772 DOI: 10.1016/j.virusres.2019.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/13/2019] [Accepted: 06/02/2019] [Indexed: 11/26/2022]
Abstract
Adjuvants are important components of vaccination strategies because they boost and accelerate the immune response. The aim of this study was to investigate the adjuvant activity of PCP-II, a polysaccharide isolated from Poria cocos, together with an inactivated rabies vaccine. The polysaccharide PCP-II was compared with the common veterinary rabies vaccine adjuvant Alhydrogel by co-administration of either adjuvant with the inactivated rabies virus rCVS-11-G to mice via the intramuscular route. Blood samples were collected to determine the virus-neutralizing antibody (VNA) titer and assess activation of B and T lymphocytes. Inguinal lymph node samples were collected, and proliferation of B lymphocytes was measured. Splenocytes were isolated, and antigen-specific cellular immune responses were evaluated by enzyme-linked immunospot and immunosorbent assays (ELISpot assay and ELISA, respectively). The results showed that PCP-II enhanced and promoted an increase in the VNA titer in the mice compared to Alhydrogel. Flow cytometry assays revealed that the polysaccharide activated more B lymphocytes in the lymph nodes and more B and T lymphocytes in the blood. Assessment of antigen-specific cellular immune responses showed that PCP-II strongly induced T lymphocyte proliferation in the spleen and high levels of cytokine secretion from splenocytes. All of these data suggest that PCP-II possesses excellent adjuvant activity and enhances both cellular and humoral immunity in mice. After examining the adjuvant activities of PCP-II in mice, dogs were immunized with rCVS-11-G together with Alhydrogel or PCP-II as an adjuvant; the control group was injected with a commercial rabies vaccine. Serum samples were collected, and the VNA titers were measured. PCP-II caused increases in the VNA titers in both the booster and single-dose immunization tests when co-administered with rCVS-11-G compared with Alhydrogel. The VNA titer of the commercial vaccine group was also significantly lower than that of the PCP-II group. These data indicate that PCP-II is an excellent candidate adjuvant for inactive rabies vaccines in the veterinary setting.
Collapse
Affiliation(s)
- Weijiao Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Nan Cheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Yuxia Wang
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Xuexing Zheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Yongkun Zhao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Hualei Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Chong Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Qiuxue Han
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Yuwei Gao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Junjie Shan
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China.
| | - Songtao Yang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.
| | - Xianzhu Xia
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| |
Collapse
|
8
|
Malyshkina A, Littwitz-Salomon E, Sutter K, Ross JA, Paschen A, Windmann S, Schimmer S, Dittmer U. Chronic retroviral infection of mice promotes tumor development, but CD137 agonist therapy restores effective tumor immune surveillance. Cancer Immunol Immunother 2019; 68:479-488. [PMID: 30635687 PMCID: PMC11028158 DOI: 10.1007/s00262-019-02300-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/06/2019] [Indexed: 12/29/2022]
Abstract
T cell responses are crucial for anti-tumor immunity. In chronic viral infections, anti-tumor T cell responses can be compromised due to various immunological mechanisms, including T cell exhaustion. To study mechanisms of anti-tumor immunity during a chronic viral infection, we made use of the well-established Friend virus (FV) mouse model. Chronically FV-infected mice are impaired in their ability to reject FBL-3 cells-a virus-induced tumor cell line of C57BL/6 origin. Here we aimed to explore therapeutic strategies to overcome the influence of T cell exhaustion during chronic viral infection, and reactivate effector CD8+ and CD4+ T cells to eliminate tumor cells. For T cell stimulation, agonistic antibodies against the tumor necrosis factor receptor (TNFR) superfamily members CD137 and CD134 were used, because they were reported to augment the cytotoxic program of T cells. αCD137 agonistic therapy, but not αCD134 agonistic therapy, resulted in FBL-3 tumor elimination in chronically FV-infected mice. CD137 stimulation significantly enhanced the cytotoxic activity of both CD4+ and CD8+ T cells, which were both required for efficient tumor control. Our study suggests that agonistic antibodies to CD137 can efficiently enhance anti-tumor immunity even in the setting of chronic viral infection, which might have promising therapeutic applications.
Collapse
Affiliation(s)
- Anna Malyshkina
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147, Essen, Germany.
| | - Elisabeth Littwitz-Salomon
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147, Essen, Germany
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147, Essen, Germany
| | - Jean Alexander Ross
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147, Essen, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sonja Windmann
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147, Essen, Germany
| | - Simone Schimmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147, Essen, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147, Essen, Germany
| |
Collapse
|
9
|
Yao S, Li Y, Zhang Q, Zhang H, Zhou L, Liao H, Zhang C, Xu M. Staphylococcal enterotoxin C2 as an adjuvant for rabies vaccine induces specific immune responses in mice. Pathog Dis 2019; 76:5025657. [PMID: 29860490 DOI: 10.1093/femspd/fty049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/29/2018] [Indexed: 01/16/2023] Open
Abstract
Rabies vaccine administration is the most effective method to prevent the occurrence of rabies disease. However, administration of rabies vaccine without adjuvant always shows low efficiency. As a member of superantigen, staphylococcal enterotoxin C2 (SEC2) non-specifically activates T-cells at extremely low concentration. It enlightens us that SEC2 may be used as an adjuvant. We carried out the experiment that the mice received twice immunization with rabies vaccine in the presence or absence of SEC2 at 1-week interval. Serum and splenocytes from immunized mice were collected to measure the level of rabies-specific-IgG and the cell that secretes IFN-γ or IL-4. The promotion of antigen-specific splenocytes proliferation was also detected. Besides, a challenge test was performed to evaluate the protective efficiency of SEC2. It was shown that mice immunized with vaccine combined with SEC2 generated more specific anti-rabies-antibodies. The results for production of IFN-γ and IL-4, as well as the proliferation of splenocytes from immunized mice indicated SEC2 promoted the specific immune responses induced by rabies vaccine. Moreover, immunization of mice with vaccine combined with SEC2 provided efficient protection against the lethal rabies exposure. Taken together, our findings indicated that SEC2 can be served as an adjuvant for rabies vaccines.
Collapse
Affiliation(s)
- Songyuan Yao
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Yongqiang Li
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Qianru Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| | - Huiwen Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| | - Libao Zhou
- Chengda Biotechnology Co. Ltd, 110179 Liaoning, China
| | - Hui Liao
- Chengda Biotechnology Co. Ltd, 110179 Liaoning, China
| | - Chenggang Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| | - Mingkai Xu
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| |
Collapse
|
10
|
Gai W, Zheng W, Wang C, Wong G, Song Y, Zheng X. Immunization with recombinant rabies virus expressing Interleukin-18 exhibits enhanced immunogenicity and protection in mice. Oncotarget 2017; 8:91505-91515. [PMID: 29207661 PMCID: PMC5710941 DOI: 10.18632/oncotarget.21065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022] Open
Abstract
Several studies have shown that interleukin-18 (IL-18) plays an important role in both innate and adaptive immune responses. In this study, we investigated the pathogenicity and immunogenicity of recombinant rabies virus expressing IL-18 (rHEP-IL18). Experimental results showed that Institute of Cancer Research (ICR) mice that received a single intramuscular immunization with rHEP-IL18 elicited the highest titers of serum neutralizing antibodies and the strongest cell-mediated immune responses to prevent the development of rabies disease, compared with immunization with the parent virus HEP-Flury. Mice inoculated with rHEP-IL18 developed significantly higher IFN-γ responses, increased percentages of CD4+ and CD8+ T-lymphocytes compared to HEP-Flury. Flow cytometry results show that rHEP-IL18 recruited more activated T- and B-cells in lymph nodes or peripheral blood, which is beneficial for virus clearance in the early stages of infection. A higher percentage of mice immunized with rHEP-IL18 survived wild-type rabies virus (RABV) challenge, compared to HEP-Flury mice. Our results show that rHEP-IL18 is promising as a novel vaccine for RABV prevention and control.
Collapse
Affiliation(s)
- Weiwei Gai
- College of Veterinary Medicine, Jilin University, Changchun, China
- School of Public Health, Shandong University, Jinan, China
| | - Wenwen Zheng
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chong Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Gary Wong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yanyan Song
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuexing Zheng
- School of Public Health, Shandong University, Jinan, China
| |
Collapse
|
11
|
Malyshkina A, Littwitz-Salomon E, Sutter K, Zelinskyy G, Windmann S, Schimmer S, Paschen A, Streeck H, Hasenkrug KJ, Dittmer U. Fas Ligand-mediated cytotoxicity of CD4+ T cells during chronic retrovirus infection. Sci Rep 2017; 7:7785. [PMID: 28798348 PMCID: PMC5552859 DOI: 10.1038/s41598-017-08578-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
CD4+ helper T cells and cytotoxic CD8+ T cells are key players for adaptive immune responses against acute infections with retroviruses. Similar to textbook knowledge the most important function of CD4+ T cells during an acute retrovirus infection seems to be their helper function for other immune cells. Whereas there was no direct anti-viral activity of CD4+ T cells during acute Friend Virus (FV) infection, they were absolutely required for the control of chronic infection. During chronic FV infection a population of activated FV-specific CD4+ T cells did not express cytotoxic molecules, but Fas Ligand that can induce Fas-induced apoptosis in target cells. Using an MHC II-restricted in vivo CTL assay we demonstrated that FV-specific CD4+ T cells indeed mediated cytotoxic effects against FV epitope peptide loaded targets. CD4 + CTL killing was also detected in FV-infected granzyme B knockout mice confirming that the exocytosis pathway was not involved. However, killing could be blocked by antibodies against FasL, which identified the Fas/FasL pathway as critical cytotoxic mechanism during chronic FV infection. Interestingly, targeting the co-stimulatory receptor CD137 with an agonistic antibody enhanced CD4+ T cell cytotoxicity. This immunotherapy may be an interesting new approach for the treatment of chronic viral infections.
Collapse
Affiliation(s)
- Anna Malyshkina
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | | | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sonja Windmann
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Simone Schimmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Annette Paschen
- Department of Dermatology, Venereology, and Allergology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hendrik Streeck
- Institute for HIV Research, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
12
|
Li SX, Barrett BS, Guo K, Santiago ML. Tetherin/BST-2: Restriction Factor or Immunomodulator? Curr HIV Res 2016; 14:235-46. [PMID: 26957198 DOI: 10.2174/1570162x14999160224102752] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/10/2015] [Accepted: 08/11/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cell-mediated immune (CMI) responses are critical for the control of HIV-1 infection and their importance was highlighted by the existence of viral proteins, particularly Vpu and Nef, that antagonize these responses. Pandemic HIV-1 Vpu counteracts Tetherin/BST-2, a host factor that could prevent the release of HIV-1 virions by tethering virions on the cell surface, but a link between Tetherin and HIV-1 CMI responses has not yet been demonstrated in vivo. In vitro, the virological and immunological impact of Tetherin-mediated accumulation of virions ranged from enhanced or diminished cell-to-cell spread to enhanced recognition by virus-specific antibodies for natural killer cellmediated lysis. However, Tetherin-restricted virions could be internalized through an endocytosis motif in the Tetherin cytoplasmic tail. METHODS Given the uncertainties on which in vitro results manifest in vivo and the dearth of knowledge on how Tetherin influences retroviral immunity, in vivo retrovirus infections in mice encoding wild-type, null and endocytosis-defective Tetherin were performed. Here, we review and highlight the results from these in vivo studies. RESULTS Current data suggests that endocytosis-defective Tetherin functions as a potent innate restriction factor. By contrast, endocytosis-competent Tetherin, the form found in most mammals including humans and the form counteracted by HIV-1 Vpu, was linked to stronger CMI responses in mice. CONCLUSION We propose that the main role of endocytosis-competent Tetherin is not to directly restrict retroviral replication, but to promote a more effective CMI response against retroviruses.
Collapse
Affiliation(s)
| | | | | | - Mario L Santiago
- Division of Infectious Diseases, University of Colorado Denver, Mail Stop B-168, 12700 E 19th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
13
|
Dietze KK, Dittmer U, Koudaimi DK, Schimmer S, Reitz M, Breloer M, Hartmann W. Filariae-Retrovirus Co-infection in Mice is Associated with Suppressed Virus-Specific IgG Immune Response and Higher Viral Loads. PLoS Negl Trop Dis 2016; 10:e0005170. [PMID: 27923052 PMCID: PMC5140070 DOI: 10.1371/journal.pntd.0005170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 11/07/2016] [Indexed: 12/22/2022] Open
Abstract
Worldwide more than 2 billion people are infected with helminths, predominantly in developing countries. Co-infections with viruses such as human immunodeficiency virus (HIV) are common due to the geographical overlap of these pathogens. Helminth and viral infections induce antagonistic cytokine responses in their hosts. Helminths shift the immune system to a type 2-dominated immune response, while viral infections skew the cytokine response towards a type 1 immune response. Moreover, chronic helminth infections are often associated with a generalized suppression of the immune system leading to prolonged parasite survival, and also to a reduced defence against unrelated pathogens. To test whether helminths affect the outcome of a viral infection we set up a filarial/retrovirus co-infection model in C57BL/6 mice. Although Friend virus (FV) infection altered the L. sigmodontis-specific immunoglobulin response towards a type I associated IgG2 isotype in co-infected mice, control of L. sigmodontis infection was not affected by a FV-superinfection. However, reciprocal control of FV infection was clearly impaired by concurrent L. sigmodontis infection. Spleen weight as an indicator of pathology and viral loads in spleen, lymph nodes (LN) and bone marrow (BM) were increased in L. sigmodontis/FV-co-infected mice compared to only FV-infected mice. Numbers of FV-specific CD8+ T cells as well as cytokine production by CD4+ and CD8+ cells were alike in co-infected and FV-infected mice. Increased viral loads in co-infected mice were associated with reduced titres of neutralising FV-specific IgG2b and IgG2c antibodies. In summary our findings suggest that helminth infection interfered with the control of retroviral infection by dampening the virus-specific neutralising antibody response. The coincidental infection of a host with two different pathogens is widespread in low-income countries. Regions where helminth infections are endemic strongly overlap with areas where the incidence of viral infections such as HIV is high. HIV is a major public health issue causing more than 1 million deaths per year. To analyse the impact of a pre-existing helminth infection on a viral infection we established a helminth/retrovirus co-infection mouse model. Mice that were first infected with Litomosoides sigmodontis and subsequently with a murine retrovirus showed a more severe course of virus infection, i.e. exaggerated splenomegaly and higher viral loads. Since different lymphocytes such as B and T cells contribute to viral control we analysed the cellular and humoral immune response. While T cell responses were similar in co-infected and virus-infected mice, we observed reduced titres of virus-specific antibodies in co-infected mice. Our results suggest that helminth infection interfered with viral control by dampening the virus-specific antibody response. The viral infection itself altered the humoral immune response against L. sigmodontis without changing the worm burden. In summary, our data highlight the importance of deworming programs or vaccines against helminths in developing countries where the incidence of helminth/HIV co-infections is high.
Collapse
Affiliation(s)
- Kirsten Katrin Dietze
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulf Dittmer
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Daniel Karim Koudaimi
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Simone Schimmer
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martina Reitz
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Minka Breloer
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Wiebke Hartmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- * E-mail:
| |
Collapse
|
14
|
Inhibition of catecholamine degradation ameliorates while chemical sympathectomy aggravates the severity of acute Friend retrovirus infection in mice. Brain Behav Immun 2016; 54:252-259. [PMID: 26880342 DOI: 10.1016/j.bbi.2016.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/29/2016] [Accepted: 02/11/2016] [Indexed: 11/21/2022] Open
Abstract
Several lines of evidence indicate that the sympathetic nervous system (SNS) might be involved in the pathogenesis and progression of retroviral infections. However, experimental data are scarce and findings inconsistent. Here, we investigated the role of the SNS during acute infection with Friend virus (FV), a pathogenic murine retrovirus that causes polyclonal proliferation of erythroid precursor cells and splenomegaly in adult mice. Experimental animals were infected with FV complex, and viral load, spleen weight, and splenic noradrenaline (NA) concentration was analyzed until 25 days post infection. Results show that FV infection caused a massive but transient depletion in splenic NA during the acute phase of the disease. At the peak of the virus-induced splenomegaly, splenic NA concentration was reduced by about 90% compared to naïve uninfected mice. Concurrently, expression of the catecholamine degrading enzymes monoamine oxidase A (MAO-A) and catechol-O-methyltransferase (COMT) was significantly upregulated in immune cells of the spleen. Pharmacological inhibition of MAO-A and COMT by the selective inhibitors clorgyline and 3,5-dinitrocatechol, respectively, efficiently blocked NA degradation and significantly reduced viral load and virus-induced splenomegaly. In contrast, chemical sympathectomy prior to FV inoculation aggravated the acute infection and extended the duration of the disease. Together these findings demonstrate that catecholamine availability at the site of viral replication is an important factor affecting the course of retroviral infections.
Collapse
|
15
|
Tas JMJ, Mesin L, Pasqual G, Targ S, Jacobsen JT, Mano YM, Chen CS, Weill JC, Reynaud CA, Browne EP, Meyer-Hermann M, Victora GD. Visualizing antibody affinity maturation in germinal centers. Science 2016; 351:1048-54. [PMID: 26912368 DOI: 10.1126/science.aad3439] [Citation(s) in RCA: 327] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/02/2016] [Indexed: 12/17/2022]
Abstract
Antibodies somatically mutate to attain high affinity in germinal centers (GCs). There, competition between B cell clones and among somatic mutants of each clone drives an increase in average affinity across the population. The extent to which higher-affinity cells eliminating competitors restricts clonal diversity is unknown. By combining multiphoton microscopy and sequencing, we show that tens to hundreds of distinct B cell clones seed each GC and that GCs lose clonal diversity at widely disparate rates. Furthermore, efficient affinity maturation can occur in the absence of homogenizing selection, ensuring that many clones can mature in parallel within the same GC. Our findings have implications for development of vaccines in which antibodies with nonimmunodominant specificities must be elicited, as is the case for HIV-1 and influenza.
Collapse
Affiliation(s)
- Jeroen M J Tas
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Luka Mesin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Giulia Pasqual
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Sasha Targ
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Johanne T Jacobsen
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.,Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Yasuko M Mano
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Casie S Chen
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Jean-Claude Weill
- Institut Necker-Enfants Malades, INSERM U1151-CNRS UMR 8253, Sorbonne Paris Cité, Université Paris Descartes, Faculté de Médecine-Site Broussais, 75014 Paris, France
| | - Claude-Agnès Reynaud
- Institut Necker-Enfants Malades, INSERM U1151-CNRS UMR 8253, Sorbonne Paris Cité, Université Paris Descartes, Faculté de Médecine-Site Broussais, 75014 Paris, France
| | - Edward P Browne
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 02142, USA.,Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Inhoffenstraβe7, 38124 Braunschweig, Germany.,Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Gabriel D Victora
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| |
Collapse
|
16
|
Zhang W, Zheng X, Cheng N, Gai W, Xue X, Wang Y, Gao Y, Shan J, Yang S, Xia X. Isatis indigotica root polysaccharides as adjuvants for an inactivated rabies virus vaccine. Int J Biol Macromol 2016; 87:7-15. [PMID: 26875535 PMCID: PMC7112441 DOI: 10.1016/j.ijbiomac.2016.02.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 10/27/2022]
Abstract
Adjuvants can enhance vaccine immunogenicity and induce long-term enhancement of immune responses. Thus, adjuvants are important for vaccine research. Polysaccharides isolated from select Chinese herbs have been demonstrated to possess various beneficial functions and excellent adjuvant abilities. In the present study, the polysaccharides IIP-A-1 and IIP-2 were isolated from Isatis indigotica root and compared with the common vaccine adjuvant aluminum hydroxide via intramuscular co-administration of inactivated rabies virus rCVS-11-G into mice. Blood was collected to determine virus neutralizing antibody (VNA) titers and B and T lymphocyte activation status. Inguinal lymph node samples were collected and used to measure B lymphocyte proliferation. Splenocytes were isolated, from which antigen-specific cellular immune responses were detected via ELISpot, ELISA and intracellular cytokine staining. The results revealed that both types of polysaccharides induce more rapid changes and higher VNA titers than aluminum hydroxide. Flow cytometry assays revealed that the polysaccharides activated more B lymphocytes in the lymph nodes and more B and T lymphocytes in the blood than aluminum hydroxide. Antigen-specific cellular immune responses showed that IIP-2 strongly induced T lymphocyte proliferation in the spleen and high levels of cytokine secretion from splenocytes, whereas aluminum hydroxide induced proliferation in only a small number of lymphocytes and the secretion of only small quantities of cytokines. Collectively, these data suggest that the polysaccharide IIP-2 exhibits excellent adjuvant activity and can enhance both cellular and humoral immunity.
Collapse
Affiliation(s)
- Weijiao Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xuexing Zheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Nan Cheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Weiwei Gai
- College of Veterinary Medicine, Jilin University, Changchun, China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xianghong Xue
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Yuxia Wang
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Yuwei Gao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Junjie Shan
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China.
| | - Songtao Yang
- College of Veterinary Medicine, Jilin University, Changchun, China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.
| | - Xianzhu Xia
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| |
Collapse
|
17
|
Jia Y, Krishnan L, Omri A. Nasal and pulmonary vaccine delivery using particulate carriers. Expert Opin Drug Deliv 2015; 12:993-1008. [PMID: 25952104 DOI: 10.1517/17425247.2015.1044435] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Many human pathogens cause respiratory illness by colonizing and invading the respiratory mucosal surfaces. Preventing infection at local sites via mucosally active vaccines is a promising and rational approach for vaccine development. However, stimulating mucosal immunity is often challenging. Particulate adjuvants that can specifically target mucosal immune cells offer a promising opportunity to stimulate local immunity at the nasal and/or pulmonary mucosal surfaces. AREAS COVERED This review analyzes the common causes of respiratory infections, the challenges in the induction of mucosal and systemic responses and current pulmonary and nasal mucosal vaccination strategies. The ability of various particulate adjuvant formulations, including lipid-based particles, polymers and other particulate systems, to be effectively utilized for mucosal vaccine delivery is discussed. EXPERT OPINION Induction of antibody and cell-mediated mucosal immunity that can effectively combat respiratory pathogens remains a challenge. Particulate delivery systems can be developed to target mucosal immune cells and effectively present antigen to evoke a rapid and long-term local immunity in the respiratory mucosa. In particular, particulate delivery systems offer the versatility of being formulated with multiple adjuvants and antigenic cargo, and can be tailored to effectively prime immune responses across the mucosal barrier. The opportunity for rational design of novel subunit particulate vaccines is emerging.
Collapse
Affiliation(s)
- Yimei Jia
- National Research Council of Canada-Human Health Therapeutics , Ottawa, Ontario K1A 0R6 , Canada +1 613 991 3210 ;
| | | | | |
Collapse
|
18
|
Venkataswamy MM, Madhusudana SN, Sanyal SS, Taj S, Belludi AY, Mani RS, Hazra N. Cellular immune response following pre-exposure and postexposure rabies vaccination by intradermal and intramuscular routes. Clin Exp Vaccine Res 2015; 4:68-74. [PMID: 25649188 PMCID: PMC4313111 DOI: 10.7774/cevr.2015.4.1.68] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 01/15/2023] Open
Abstract
Purpose Immunization against rabies in humans induces protective neutralizing antibodies; however, the induction of type 1 or type 2 cytokine mediated cellular immune responses following rabies vaccination is not understood. Hence, the present study investigated cellular cytokine responses in vaccinated individuals. Materials and Methods The study groups included healthy rabies antigen naive controls (n=10), individuals who received intradermal primary (n=10) or booster pre-exposure vaccination (n=20) and subjects who received postexposure rabies vaccination either by intradermal (n=18) or intramuscular (n=20) routes. The antigen specific cellular responses were analyzed by stimulating peripheral blood mononuclear cells with a rabies vaccine antigen in the interferon-γ (IFN-γ) and interleukin-4 (IL-4) enzyme-linked immunospot (ELISpot) assay. These responses were compared to the rabies virus neutralizing antibody (RVNA) titers that were measured by rapid fluorescent focus inhibition test. Results We observed that cellular and humoral immune responses to primary intradermal rabies vaccination could be greatly enhanced by a booster vaccine; and both type 1 and type 2 cytokine responses were significantly elevated. The magnitude of type 1 and type 2 cytokine responses did not differ significantly among the intramuscular and intradermal routes of postexposure vaccination. The number of cells producing IFN-γ and IL-4 correlated significantly with the levels of RVNA. Conclusion Both type 1 and type 2 cellular cytokine responses are strongly induced after rabies vaccination and directly correlate with levels of RVNA titers. The neutralizing antibody as well as the type 1 and type 2 cytokine responses may be important for vaccine induced protective responses against rabies.
Collapse
Affiliation(s)
| | | | - Sampada Sudarshan Sanyal
- Deptartment of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Shaheen Taj
- Deptartment of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Ashwin Yajaman Belludi
- Deptartment of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Reeta Subramaniam Mani
- Deptartment of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Nandita Hazra
- Deptartment of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, India
| |
Collapse
|
19
|
Knuschke T, Bayer W, Rotan O, Sokolova V, Wadwa M, Kirschning CJ, Hansen W, Dittmer U, Epple M, Buer J, Westendorf AM. Prophylactic and therapeutic vaccination with a nanoparticle-based peptide vaccine induces efficient protective immunity during acute and chronic retroviral infection. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1787-98. [DOI: 10.1016/j.nano.2014.06.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/13/2014] [Indexed: 01/31/2023]
|
20
|
Endogenous retrovirus induces leukemia in a xenograft mouse model for primary myelofibrosis. Proc Natl Acad Sci U S A 2014; 111:8595-600. [PMID: 24912157 DOI: 10.1073/pnas.1401215111] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The compound immunodeficiencies in nonobese diabetic (NOD) inbred mice homozygous for the Prkdc(scid) and Il2rg(null) alleles (NSG mice) permit engraftment of a wide-range of primary human cells, enabling sophisticated modeling of human disease. In studies designed to define neoplastic stem cells of primary myelofibrosis (PMF), a myeloproliferative neoplasm characterized by profound disruption of the hematopoietic microenvironment, we observed a high frequency of acute myeloid leukemia (AML) in NSG mice. AML was of mouse origin, confined to PMF-xenografted mice, and contained multiple clonal integrations of ecotropic murine leukemia virus (E-MuLV). Significantly, MuLV replication was not only observed in diseased mice, but also in nontreated NSG controls. Furthermore, in addition to the single ecotropic endogenous retrovirus (eERV) located on chromosome 11 (Emv30) in the NOD genome, multiple de novo germ-line eERV integrations were observed in mice from each of four independent NSG mouse colonies. Analysis confirmed that E-MuLV originated from the Emv30 provirus and that recombination events were not necessary for virus replication or AML induction. Pathogenicity is thus likely attributable to PMF-mediated paracrine stimulation of mouse myeloid cells, which serve as targets for retroviral infection and transformation, as evidenced by integration into the Evi1 locus, a hotspot for retroviral-induced myeloid leukemia. This study thus corroborates a role of paracrine stimulation in PMF disease progression, underlines the importance of target cell type and numbers in MuLV-induced disease, and mandates awareness of replicating MuLV in NOD immunodeficient mice, which can significantly influence experimental results and their interpretation.
Collapse
|
21
|
Messer RJ, Lavender KJ, Hasenkrug KJ. Mice of the resistant H-2(b) haplotype mount broad CD4(+) T cell responses against 9 distinct Friend virus epitopes. Virology 2014; 456-457:139-44. [PMID: 24889233 DOI: 10.1016/j.virol.2014.03.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/10/2014] [Accepted: 03/12/2014] [Indexed: 12/16/2022]
Abstract
To date, only a single Friend virus (FV) peptide recognized by CD4(+) T cells in FV-infected mice of the resistant H-2(b) haplotype has been described. To more thoroughly examine the repertoire of CD4(+) T cell responses in H-2(b) mice infected with this retrovirus, 18mer peptides spanning the FV gag, pol, and env coding regions with 11mer overlaps were synthesized. The peptides were then used to stimulate whole splenocytes and purified CD4(+) T cells from FV-infected mice in an IFNγ ELISPOT assay. Nine new CD4(+) T cell epitopes were identified, 3 encoded by gag, 1 by pol, and 5 by env. The high resistance of H-2(b) mice could be related to this very broad CD4(+) T cell response against multiple peptides during FV infection.
Collapse
Affiliation(s)
- Ronald J Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT 59840, USA
| | - Kerry J Lavender
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT 59840, USA
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT 59840, USA.
| |
Collapse
|
22
|
Li SX, Barrett BS, Harper MS, Heilman KJ, Halemano K, Steele AK, Guo K, Silverman RH, Santiago ML. Ribonuclease L is not critical for innate restriction and adaptive immunity against Friend retrovirus infection. Virology 2013; 443:134-42. [PMID: 23725696 DOI: 10.1016/j.virol.2013.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 04/17/2013] [Accepted: 05/03/2013] [Indexed: 01/01/2023]
Abstract
Ribonuclease L (RNase L) is a type I interferon regulated factor that can significantly inhibit retroviruses in vitro and may activate cytoplasmic sensing pathways to augment adaptive immunity. However, the antiretroviral activity of RNase L remains to be validated in vivo. We investigated the role of RNaseL in counteracting Friend retrovirus (FV) infection relative to a well-described restriction factor, Apobec3. C57BL/6 wild-type (WT) and RNaseL knock-out (KO) mice exhibited similar acute FV infection levels despite significant transcriptional induction of oligoadenylate synthetase 1, which produces activators of RNase L. Apobec3 KO mice showed higher FV infection levels relative to WT mice, but deletion of RNaseL in Apobec3 KO mice did not augment FV infection. Moreover, RNaseL did not influence FV-specific IgG responses and recovery from viremia by 28 days post-infection. The results suggest that RNase L is not an evolutionarily-conserved host defense mechanism to counteract retroviruses in vivo.
Collapse
Affiliation(s)
- Sam X Li
- Division of Infectious Diseases, University of Colorado Denver, CO 80045, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Walton S, Mandaric S, Oxenius A. CD4 T cell responses in latent and chronic viral infections. Front Immunol 2013; 4:105. [PMID: 23717308 PMCID: PMC3651995 DOI: 10.3389/fimmu.2013.00105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/22/2013] [Indexed: 12/24/2022] Open
Abstract
The spectrum of tasks which is fulfilled by CD4 T cells in the setting of viral infections is large, ranging from support of CD8 T cells and humoral immunity to exertion of direct antiviral effector functions. While our knowledge about the differentiation pathways, plasticity, and memory of CD4 T cell responses upon acute infections or immunizations has significantly increased during the past years, much less is still known about CD4 T cell differentiation and their beneficial or pathological functions during persistent viral infections. In this review we summarize current knowledge about the differentiation, direct or indirect antiviral effector functions, and the regulation of virus-specific CD4 T cells in the setting of persistent latent or active chronic viral infections with a particular emphasis on herpes virus infections for the former and chronic lymphocytic choriomeningitis virus infection for the latter.
Collapse
Affiliation(s)
- Senta Walton
- Department of Microbiology and Immunology, School of Pathology and Laboratory Medicine, University of Western Australia Nedlands, WA, Australia
| | | | | |
Collapse
|
24
|
CD4+ T cells develop antiretroviral cytotoxic activity in the absence of regulatory T cells and CD8+ T cells. J Virol 2013; 87:6306-13. [PMID: 23536666 DOI: 10.1128/jvi.00432-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Conventional CD4(+) T cells play an important role in viral immunity. In most virus infections, they provide essential help for antiviral B and T cell responses. In chronic infections, including HIV infection, an expansion of regulatory T cells (Tregs) has been demonstrated, which can suppress virus-specific CD4(+) T cell responses in vitro. However, the suppressive activity of Tregs on effector CD4(+) T cells in retroviral infection is less well documented in vivo. We took advantage of a transgenic mouse in which Tregs can be selectively depleted to determine the influence of such cells on retrovirus-specific CD4(+) T cell responses during an ongoing infection. Mice were infected with Friend retrovirus (FV), and Tregs were depleted during the acute phase of the infection. In nondepleted mice, activated CD4(+) T cells produced Th1-type cytokines but did not exhibit any antiviral cytotoxicity as determined in a major histocompatibility complex (MHC) class II-restricted in vivo cytotoxic T lymphocyte (CTL) assay. Depletion of Tregs significantly increased the numbers of virus-specific CD4(+) T cells and improved their cytokine production, whereas it induced only very little CD4(+) T cell cytotoxicity. However, after dual depletion of Tregs and CD8(+) T cells, conventional CD4(+) T cells developed significant cytotoxic activity against FV epitope-labeled target cells in vivo and contributed to the control of virus replication. Thus, both Tregs and CD8(+) T cells influence the cytotoxic activity of conventional CD4(+) T cells during an acute retroviral infection.
Collapse
|