1
|
Chakraborty C, Lo YH, Bhattacharya M, Das A, Wen ZH. Looking beyond the origin of SARS-CoV-2: Significant strategic aspects during the five-year journey of COVID-19 vaccine development. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102527. [PMID: 40291378 PMCID: PMC12032352 DOI: 10.1016/j.omtn.2025.102527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
It has been five years since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and we are also approaching the five-year mark of the COVID-19 pandemic. The vaccine is a significant weapon in combating infectious diseases like SARS-CoV-2. Several vaccines were developed against SARS-CoV-2, and they demonstrated efficacy and safety during these five years. The rapid development of multiple next-generation vaccine candidates in different platforms with very little time is the success story of the vaccine development endeavor. This remarkable success of rapid vaccine development is a new paradigm for fast vaccine development that might help develop infectious diseases and fight against the pandemic. With the completion of five years since the beginning of SARS-CoV-2 origin, we are looking back on the five years and reviewing the milestones, vaccine platforms, animal models, clinical trials, successful collaborations, vaccine safety, real-world effectiveness, and challenges. Lessons learned during these five years will help us respond to public health emergencies and to fight the battle against future pandemics.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 81342, Taiwan
- Department of Nursing, Meiho University, Neipu Township, Pingtung County 91200, Taiwan
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020, India
| | - Arpita Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, #70 Lien-Hai Road, Kaohsiung 804201, Taiwan
- National Museum of Marine Biology & Aquarium, # 2 Houwan Road, Checheng, Pingtung 94450, Taiwan
| |
Collapse
|
2
|
Ying-Fen L, Chun S, Jun Y, Shuai-Bo H, Yu-Jie C, Gui-Lan W, Kai-Gong W, Chun-Lan S, Er-Peng Z, Zhen-Tao C. Recombinant adenovirus expressing pdhβ-pdhD fusion protein produces robust immune responses and partial protection against Mycoplasma Synoviae challenge in chickens. Poult Sci 2025; 104:105185. [PMID: 40315580 DOI: 10.1016/j.psj.2025.105185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
Mycoplasma synoviae (MS) stands as a pivotal pathogen, responsible for triggering arthritis and airsacculitis in both chickens and turkeys. Given the pressing need for safe and efficacious vaccine candidates, we engineered recombinant adenoviruses expressing a fusion antigen. This antigen consisted of the pyruvate dehydrogenase E1 subunit beta (pdhβ) and dihydrolipoyl dehydrogenase (pdhD) of MS. We then systematically evaluated the immune effect and protective efficacy of these recombinant adenoviruses against MS challenge in a chicken model. Our results demonstrated the successful construction of recombinant adenoviruses rAd-pdhβ, rAd-pdhD, and rAd-pdhβ-pdhD. The pdhβ, pdhD, and pdhβ-pdhD proteins were efficiently expressed in cells infected with the respective recombinant adenoviruses. Animal experiments further revealed that vaccination with recombinant adenoviruses rAd-pdhβ, rAd-pdhD, and rAd-pdhβ-pdhD elicited significant specific humoral and cellular immune responses (P < 0.05). Notably, rAd-pdhβ-pdhD exhibited superior immunogenicity compared to rAd-pdhβ and rAd-pdhD. Moreover, all three recombinant adenovirus vaccine candidates conferred partial protection to chickens against MS challenge. They effectively alleviated MS-induced footpad and joint swelling, as well as inflammation. Among them, rAd-pdhβ-pdhD demonstrated a better protective effect. In conclusion, vaccination with recombinant adenoviruses rAd-pdhβ, rAd-pdhD, and rAd-pdhβ-pdhD can evoke immune responses and provide partial protection against MS in chickens. In particular, rAd-pdhβ-pdhD holds greater potential as a vaccine candidate against MS.
Collapse
Affiliation(s)
- Li Ying-Fen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Song Chun
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Yue Jun
- Guizhou Animal Disease Prevention and Control Center, Guiyang 550000, Guizhou, PR China
| | - Han Shuai-Bo
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Chen Yu-Jie
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Wen Gui-Lan
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Wang Kai-Gong
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Shan Chun-Lan
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Zhu Er-Peng
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Cheng Zhen-Tao
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, PR China.
| |
Collapse
|
3
|
Xu M, Yang Z, Yang N, Li H, Ma H, Yi J, Hou H, Han F, Ma Z, Chen C. Development and Immunogenicity Study of Subunit Vaccines Based on Spike Proteins of Porcine Epidemic Diarrhea Virus and Porcine Transmissible Gastroenteritis Virus. Vet Sci 2025; 12:106. [PMID: 40005866 PMCID: PMC11860644 DOI: 10.3390/vetsci12020106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/15/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) are responsible for significant economic losses in the swine industry. The S1 proteins of these viruses serve as key targets for vaccine development. In this study, prokaryotic expression vectors for pCZN1-PEDV S1, pCZN1-TGEV S1, and pCZN1-PEDV S1-TGEV S1 were constructed. The corresponding proteins were expressed, purified, and used to prepare monovalent, bivalent, and mixed (PEDV S1 + TGEV S1) vaccines. Kunming (KM) mice were immunized with subunit vaccines, with PBS as the negative control (NC) and a commercial inactivated vaccine as the positive control (PC). Immune responses, including specific antibody (IgG, IgG1, IgG2a) levels, virus neutralization, and IFN-γ production, were evaluated. All vaccines induced high levels of specific IgG, IgG1, and IgG2a antibodies. At weeks 2 and 8, the PEDV S1 + TGEV S1 vaccine induced significantly higher levels of specific IgG and IgG1 compared to the PC (p < 0.001). The PEDV S1 vaccine also induced significantly higher specific IgG2a levels than the PC at week 4 (p < 0.0001). Virus neutralization assays demonstrated that the subunit vaccines induced neutralizing antibody levels comparable to or exceeding those of the PC. Furthermore, IFN-γ levels were significantly elevated in all vaccinated groups compared to the NC (p < 0.0001), indicating a robust immune response. These results suggest that the subunit vaccines are promising candidates for the safe and effective control of both PEDV and TGEV infections.
Collapse
Affiliation(s)
- Mingguo Xu
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Zhonglian Yang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Ningning Yang
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang 464000, China;
| | - Honghuan Li
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Hailong Ma
- Department of Biotechnology, Linxia Modern Career Academy, Linxia 731100, China;
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Huilin Hou
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Fangfang Han
- The College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China;
| | - Zhongchen Ma
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (M.X.); (Z.Y.); (H.L.); (J.Y.); (H.H.)
| |
Collapse
|
4
|
Saini I, Joshi J, Kaur S. Unleashing the role of potential adjuvants in leishmaniasis. Int J Pharm 2025; 669:125077. [PMID: 39675537 DOI: 10.1016/j.ijpharm.2024.125077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Leishmaniasis is amongst one of the most neglected tropical disease, caused by an intracellular protozoan of genus Leishmania. Currently, the most promising strategy to combat leishmaniasis, relies on chemotherapy but the toxicity and increasing resistance of the standard drugs, presses the demand for new alternatives. Immunization is arguably the best strategy for cure because an individual once infected becomes immune to the disease. Yet, there is no efficient vaccine capable of providing enduring immunity against the parasite. Achieving the goal of developing highly efficacious and durable vaccine is limited due to lack of an appropriate adjuvant. Adjuvants are recognized as 'immune potentiators' which redirect or amplify the immune response. A number of adjuvants like alum, MPL-A, CpG ODN, GLA-SE, imiquimod, saponins etc. have been used in combination with various classes of Leishmania antigens. However, only few have reached clinical trials. Thus, the choice of an adjuvant is critically dependent on many factors such as the route of administration, the nature of antigen, formulation, the type of required immune response, their mode of action and the immunization schedule. This review provides an updated status on the types of adjuvants used in leishmaniasis so far and their mechanism of action if known.
Collapse
Affiliation(s)
- Isha Saini
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India
| | - Jyoti Joshi
- Goswami Ganesh Dutta Sanatan Dharma College, Sector-32C, Chandigarh, India
| | - Sukhbir Kaur
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India.
| |
Collapse
|
5
|
Aldahlawi AM, Zaher KSA. Dendritic Cell-Based Immunity: Screening of Dendritic Cell Subsets in Breast Cancer-Bearing Mice. J Microsc Ultrastruct 2023; 11:150-160. [PMID: 38025181 PMCID: PMC10679829 DOI: 10.4103/jmau.jmau_85_22] [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: 09/19/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/19/2023] Open
Abstract
Background Breast cancer (BC) is the most devastating disease, particularly the lethal invasive form. It is the most underlying cause of death among women worldwide. The expansion of BC is controlled by a variety of alterations in the tumor cells themselves, in addition to the state of the immune system, which has a direct influence on the tumor microenvironment. Numerous receptors expressed by T-cells interact with ligands on antigen-presenting cells to provide activation signals results in mounting effector anti-tumor T-cell responses. On the other hand, there is a dearth of information about the actual interactions and reactions of T-cells and dendritic cells (DCs) all through the progression of tumor development. Aim Immune system response against BC was investigated through tumor induction in mice. The size and volume of the tumor were calculated. Moreover, the phenotypical profile of T-cells and DCs from lymph nodes (LN) and spleens of BC-bearing mice was investigated. In addition, the levels of Transforming growth factor-β, Interferon-gamma (IFN-γ), Interleukin IL-2, IL-10, IL-4, IL-12, and tumor necrosis factor (TNF)-α were determined. Materials and Methods MDA231 cells were utilized to induce BC in 30 white BALB/C mice, whereas the other 30 mice acted as healthy controls and were not treated with any cancer-causing agents. The impact of malignancy was evaluated using flow cytometry based on the marking surface molecules, as well as the titer of specific cytokines of the mice's LN culture using the ELISA method. These cytokines included transforming growth factor-β (TGF-β), IFN-γ, IL-2, IL -10, IL -4, IL -12, and TNF-α. Results The findings showed that the maturation of DCs was inhibited, followed by an accumulation of immature DCs. These immature DCs increase the release of TGF-β and cytokines like IL-10 and inhibit the release of IFN-γ and IL-12 in the culture supernatant of nodal lymph and spleen suspension of BC-bearing mice compared to control. In addition, there was a low expression of CD80 and CD86 on DCs, which indicates a low maturation process. Conclusion According to the findings, the tumor microenvironment may have been responsible for preventing the maturation of DCs. This, in turn, weakened the immune response and facilitated the ability of the tumor to proliferate. Furthermore, the tumor microenvironment increased the number of immature DCs by inhibiting their stimulation by overexpression of TGF-β-produced by regulatory T lymphocytes and stimulation of tumor cells. In addition, the tumor microenvironment stimulated the secretion of cytokines such as IL-10, and CD4 and decreased the secretion of IFN-γ-and IL-12 in tumor-induced mice cultured LN and spleen.
Collapse
Affiliation(s)
- Alia M Aldahlawi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Kawther Sayed Ali Zaher
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| |
Collapse
|
6
|
Ge P, Li H, Ya X, Xu Y, Ma L, He Q, Wang R, Liu Z, Zhang Q, Zhang Y, Wang W, Zhang D, Zhao J. Single-cell atlas reveals different immune environments between stable and vulnerable atherosclerotic plaques. Front Immunol 2023; 13:1085468. [PMID: 36741406 PMCID: PMC9889979 DOI: 10.3389/fimmu.2022.1085468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction Regardless of the degree of stenosis, vulnerable plaque is an important cause of ischemic stroke and thrombotic complications. The changes of the immune microenvironment within plaques seem to be an important factor affecting the characteristics of the plaque. However, the differences of immune microenvironment between stable and vulnerable plaques were remained unknown. Methods In this study, RNA-sequencing was performed on superficial temporal arteries from 5 traumatic patients and plaques from 3 atherosclerotic patients to preliminary identify the key immune response processes in plaques. Mass cytometry (CyTOF) technology was used to explore differences in immune composition between 9 vulnerable plaques and 12 stable plaques. Finally, immunofluorescence technique was used to validate our findings in the previous analysis. Results Our results showed that more CD86+CD68+ M1 pro-inflammatory macrophages were found in vulnerable plaques, while CD4+T memory cells were mainly found in stable plaques. In addition, a CD11c+ subset of CD4+T cells with higher IFN-r secretion was found within the vulnerable plaque. In two subsets of B cells, CD19+CD20-B cells in vulnerable plaques secreted more TNF-a and IL-6, while CD19-CD20+B cells expressed more PD-1 molecules. Conclusion In conclusion, our study suggested that M1-like macrophages are the major cell subset affecting plaque stability, while functional B cells may also contribute to plaque stability.
Collapse
Affiliation(s)
- Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiaolong Ya
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yiqiao Xu
- Capital Medical University, Beijing, China
| | - Long Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zechen Liu
- Department of Biostatistics, Harvard School of Public Health, Huntington Avenue, Boston, MA, United States
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China,*Correspondence: Wenjing Wang, ; Dong Zhang, ; Jizong Zhao,
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,Department of Neurosurgery, Beijing Hospital, Beijing, China,*Correspondence: Wenjing Wang, ; Dong Zhang, ; Jizong Zhao,
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,*Correspondence: Wenjing Wang, ; Dong Zhang, ; Jizong Zhao,
| |
Collapse
|
7
|
The dual role of cytokine responses to Chlamydia trachomatis infection in host pathogen crosstalk. Microb Pathog 2022; 173:105812. [DOI: 10.1016/j.micpath.2022.105812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
|
8
|
Balmert SC, Ghozloujeh ZG, Carey CD, Williams LH, Zhang J, Shahi P, Amer M, Sumpter TL, Erdos G, Korkmaz E, Falo LD. A microarray patch SARS-CoV-2 vaccine induces sustained antibody responses and polyfunctional cellular immunity. iScience 2022; 25:105045. [PMID: 36062075 PMCID: PMC9425707 DOI: 10.1016/j.isci.2022.105045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/19/2022] [Accepted: 08/25/2022] [Indexed: 12/01/2022] Open
Abstract
Sustainable global immunization campaigns against COVID-19 and other emerging infectious diseases require effective, broadly deployable vaccines. Here, we report a dissolvable microarray patch (MAP) SARS-CoV-2 vaccine that targets the immunoresponsive skin microenvironment, enabling efficacious needle-free immunization. Multicomponent MAPs delivering both SARS-CoV-2 S1 subunit antigen and the TLR3 agonist Poly(I:C) induce robust antibody and cellular immune responses systemically and in the respiratory mucosa. MAP vaccine-induced antibodies bind S1 and the SARS-CoV-2 receptor-binding domain, efficiently neutralize the virus, and persist at high levels for more than a year. The MAP platform reduces systemic toxicity of the delivered adjuvant and maintains vaccine stability without refrigeration. When applied to human skin, MAP vaccines activate skin-derived migratory antigen-presenting cells, supporting the feasibility of human translation. Ultimately, this shelf-stable MAP vaccine improves immunogenicity and safety compared to traditional intramuscular vaccines and offers an attractive alternative for global immunization efforts against a range of infectious pathogens.
Collapse
Affiliation(s)
- Stephen C. Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Li’an H. Williams
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Preeti Shahi
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Maher Amer
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L. Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Louis D. Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
9
|
Zhang S, Pang K, Feng X, Zeng Y. Transcriptomic data exploration of consensus genes and molecular mechanisms between chronic obstructive pulmonary disease and lung adenocarcinoma. Sci Rep 2022; 12:13214. [PMID: 35918384 PMCID: PMC9345949 DOI: 10.1038/s41598-022-17552-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/27/2022] [Indexed: 11/09/2022] Open
Abstract
Most current research has focused on chronic obstructive pulmonary disease (COPD) and lung adenocarcinoma (LUAD) alone; however, it is important to understand the complex mechanism of COPD progression to LUAD. This study is the first to explore the unique and jointly molecular mechanisms in the pathogenesis of COPD and LUAD across several datasets based on a variety of analysis methods. We used weighted correlation network analysis to search hub genes in two datasets from public databases: GSE10072 and GSE76925. We explored the unique and jointly molecular mechanistic signatures of the two diseases in pathogenesis through enrichment analysis, immune infiltration analysis, and therapeutic targets analysis. Finally, the results were confirmed using real-time quantitative reverse transcription PCR. Fifteen hub genes were identified: GPI, EZH2, EFNA4, CFB, ENO1, SH3PXD2B, SELL, CORIN, MAD2L1, CENPF, TOP2A, ASPM, IGFBP2, CDKN2A, and ELF3. For the first time, SELL, CORIN, GPI, and EFNA4 were found to play a role in the etiology of COPD and LUAD. The LUAD genes identified were primarily involved in the cell cycle and DNA replication processes; COPD genes we found were related to ubiquitin-mediated proteolysis, ribosome, and T/B-cell receptor signaling pathways. The tumor microenvironment of LUAD pathogenesis was influenced by CD4 + T cells, type 1 regulatory T cells, and T helper 1 cells. T follicular helper cells, natural killer T cells, and B cells all impact the immunological inflammation in COPD. The results of drug targets analysis suggest that cisplatin and tretinoin, as well as bortezomib and metformin may be potential targeted therapy for patients with COPD combined LUAD. These signatures may be provided a new direction for developing early interventions and treatments to improve the prognosis of COPD and LUAD.
Collapse
Affiliation(s)
- Siyu Zhang
- Department of Respiratory Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 39 Yanhu Avenue, Wuchang District, Wuhan, 430000, Hubei, China
| | - Kun Pang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xinyu Feng
- Department of Respiratory Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 39 Yanhu Avenue, Wuchang District, Wuhan, 430000, Hubei, China
| | - Yulan Zeng
- Department of Respiratory Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 39 Yanhu Avenue, Wuchang District, Wuhan, 430000, Hubei, China.
| |
Collapse
|
10
|
Jia Z, Pan X, Zhi W, Chen H, Bai B, Ma C, Ma D. Probiotics Surface-Delivering Fiber2 Protein of Fowl Adenovirus 4 Stimulate Protective Immunity Against Hepatitis-Hydropericardium Syndrome in Chickens. Front Immunol 2022; 13:919100. [PMID: 35837390 PMCID: PMC9273852 DOI: 10.3389/fimmu.2022.919100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/27/2022] [Indexed: 12/19/2022] Open
Abstract
Background and ObjectivesHepatitis-hydropericardium syndrome (HHS) caused by Fowl adenoviruses serotype 4 (FAdV-4) leads to severe economic losses to the poultry industry. Although various vaccines are available, vaccines that effectively stimulate intestinal mucosal immunity are still deficient. In the present study, novel probiotics that surface-deliver Fiber2 protein, the major virulence determiner and efficient immunogen for FAdV-4, were explored to prevent this fecal–oral-transmitted virus, and the induced protective immunity was evaluated after oral immunization.MethodsThe probiotic Enterococcus faecalis strain MDXEF-1 and Lactococcus lactis NZ9000 were used as host strains to deliver surface-anchoring Fiber2 protein of FAdV-4. Then the constructed live recombinant bacteria were orally vaccinated thrice with chickens at intervals of 2 weeks. Following each immunization, immunoglobulin G (IgG) in sera, secretory immunoglobulin A (sIgA) in jejunum lavage, immune-related cytokines, and T-cell proliferation were detected. Following challenge with the highly virulent FAdV-4, the protective effects of the probiotics surface-delivering Fiber2 protein were evaluated by verifying inflammatory factors, viral load, liver function, and survival rate.ResultsThe results demonstrated that probiotics surface-delivering Fiber2 protein stimulated humoral and intestinal mucosal immune responses in chickens, shown by high levels of sIgA and IgG antibodies, substantial rise in mRNA levels of cytokines, increased proliferative ability of T cells in peripheral blood, improved liver function, and reduced viral load in liver. Accordingly, adequate protection against homologous challenges and a significant increase in the overall survival rate were observed. Notably, chickens orally immunized with E. faecalis/DCpep-Fiber2-CWA were completely protected from the FAdV-4 challenge, which is better than L. lactis/DCpep-Fiber2-CWA.ConclusionThe recombinant probiotics surface-expressing Fiber2 protein could evoke remarkable humoral and cellular immune responses, relieve injury, and functionally damage target organs. The current study indicates a promising method used for preventing FAdV-4 infection in chickens.
Collapse
Affiliation(s)
- Zhipeng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinghui Pan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wenjing Zhi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hang Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Bingrong Bai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chunli Ma
- College of Food Science, Northeast Agricultural University, Harbin, China
- *Correspondence: Chunli Ma, ; Dexing Ma,
| | - Dexing Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Experimental Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
- *Correspondence: Chunli Ma, ; Dexing Ma,
| |
Collapse
|
11
|
Sankaradoss A, Jagtap S, Nazir J, Moula SE, Modak A, Fialho J, Iyer M, Shastri JS, Dias M, Gadepalli R, Aggarwal A, Vedpathak M, Agrawal S, Pandit A, Nisheetha A, Kumar A, Bordoloi M, Shafi M, Shelar B, Balachandra SS, Damodar T, Masika MM, Mwaura P, Anzala O, Muthumani K, Sowdhamini R, Medigeshi GR, Roy R, Pattabiraman C, Krishna S, Sreekumar E. Immune profile and responses of a novel dengue DNA vaccine encoding an EDIII-NS1 consensus design based on Indo-African sequences. Mol Ther 2022; 30:2058-2077. [PMID: 34999210 PMCID: PMC8736276 DOI: 10.1016/j.ymthe.2022.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/24/2021] [Accepted: 01/05/2022] [Indexed: 12/30/2022] Open
Abstract
The ongoing COVID-19 pandemic highlights the need to tackle viral variants, expand the number of antigens, and assess diverse delivery systems for vaccines against emerging viruses. In the present study, a DNA vaccine candidate was generated by combining in tandem envelope protein domain III (EDIII) of dengue virus serotypes 1-4 and a dengue virus (DENV)-2 non-structural protein 1 (NS1) protein-coding region. Each domain was designed as a serotype-specific consensus coding sequence derived from different genotypes based on the whole genome sequencing of clinical isolates in India and complemented with data from Africa. This sequence was further optimized for protein expression. In silico structural analysis of the EDIII consensus sequence revealed that epitopes are structurally conserved and immunogenic. The vaccination of mice with this construct induced pan-serotype neutralizing antibodies and antigen-specific T cell responses. Assaying intracellular interferon (IFN)-γ staining, immunoglobulin IgG2(a/c)/IgG1 ratios, and immune gene profiling suggests a strong Th1-dominant immune response. Finally, the passive transfer of immune sera protected AG129 mice challenged with a virulent, non-mouse-adapted DENV-2 strain. Our findings collectively suggest an alternative strategy for dengue vaccine design by offering a novel vaccine candidate with a possible broad-spectrum protection and a successful clinical translation either as a stand alone or in a mix and match strategy.
Collapse
Affiliation(s)
- Arun Sankaradoss
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India.
| | - Suraj Jagtap
- Department of Chemical Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Junaid Nazir
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Shefta E Moula
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Ayan Modak
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala 695014, India
| | - Joshuah Fialho
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Meenakshi Iyer
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Jayanthi S Shastri
- Department of Microbiology, T.N.Medical College & B.y.L.Nair Hospital, Mumbai 400008, India
| | - Mary Dias
- Division of Infectious Disease, St. John's Medical College and Hospital, Bangalore 560034, India
| | - Ravisekhar Gadepalli
- Department of Microbiology, All India Institute of Medical Sciences, Jodhpur 342005, India
| | - Alisha Aggarwal
- Department of Microbiology, All India Institute of Medical Sciences, Jodhpur 342005, India
| | - Manoj Vedpathak
- Department of Microbiology, T.N.Medical College & B.y.L.Nair Hospital, Mumbai 400008, India
| | - Sachee Agrawal
- Department of Microbiology, T.N.Medical College & B.y.L.Nair Hospital, Mumbai 400008, India
| | - Awadhesh Pandit
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Amul Nisheetha
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Anuj Kumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Mahasweta Bordoloi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Mohamed Shafi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Bhagyashree Shelar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Swathi S Balachandra
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Tina Damodar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Moses Muia Masika
- KAVI Institute of Clinical Research, University of Nairobi, Nairobi 19676-00202, Kenya
| | - Patrick Mwaura
- KAVI Institute of Clinical Research, University of Nairobi, Nairobi 19676-00202, Kenya
| | - Omu Anzala
- KAVI Institute of Clinical Research, University of Nairobi, Nairobi 19676-00202, Kenya
| | - Kar Muthumani
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - Ramanathan Sowdhamini
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | | | - Rahul Roy
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India; Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Chitra Pattabiraman
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Sudhir Krishna
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India; School of Interdisciplinary Life Sciences, Indian Institute of Technology Goa, Ponda 404401, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala 695014, India.
| |
Collapse
|
12
|
Portilho AI, Gimenes Lima G, De Gaspari E. Enzyme-Linked Immunosorbent Assay: An Adaptable Methodology to Study SARS-CoV-2 Humoral and Cellular Immune Responses. J Clin Med 2022; 11:1503. [PMID: 35329828 PMCID: PMC8948777 DOI: 10.3390/jcm11061503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023] Open
Abstract
The Enzyme-Linked Immunosorbent Assay is a versatile technique, which can be used for several applications. It has enormously contributed to the study of infectious diseases. This review highlights how this methodology supported the science conducted in COVID-19 pandemics, allowing scientists to better understand the immune response against SARS-CoV-2. ELISA can be modified to assess the functionality of antibodies, as avidity and neutralization, respectively by the standardization of avidity-ELISA and surrogate-neutralization methods. Cellular immunity can also be studied using this assay. Products secreted by cells, like proteins and cytokines, can be studied by ELISA or its derivative Enzyme-linked immunospot (ELISpot) assay. ELISA and ELISA-based methods aided the area of immunology against infectious diseases and is still relevant, for example, as a promising approach to study the differences between natural and vaccine-induced immune responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, Sao Paulo 01246-902, SP, Brazil; (A.I.P.); (G.G.L.)
- Graduate Program Interunits in Biotechnology, University of Sao Paulo, Sao Paulo 05508-900, SP, Brazil
| | - Gabrielle Gimenes Lima
- Immunology Center, Adolfo Lutz Institute, Sao Paulo 01246-902, SP, Brazil; (A.I.P.); (G.G.L.)
- Graduate Program Interunits in Biotechnology, University of Sao Paulo, Sao Paulo 05508-900, SP, Brazil
| | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, Sao Paulo 01246-902, SP, Brazil; (A.I.P.); (G.G.L.)
- Graduate Program Interunits in Biotechnology, University of Sao Paulo, Sao Paulo 05508-900, SP, Brazil
| |
Collapse
|
13
|
Yin D, He L, Zhu E, Fang T, Yue J, Wen M, Wang K, Cheng Z. A fowl adenovirus serotype 4 (FAdV-4) Fiber2 subunit vaccine candidate provides complete protection against challenge with virulent FAdV-4 strain in chickens. Vet Microbiol 2021; 263:109250. [PMID: 34649009 DOI: 10.1016/j.vetmic.2021.109250] [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: 08/06/2021] [Accepted: 10/02/2021] [Indexed: 12/21/2022]
Abstract
Hypervirulent fowl adenovirus serotype 4 (FAdV-4)-induced hepatitis-hydropericardium syndrome (HHS) with high mortality causes huge economic losses to the poultry industry worldwide. However, commercially available vaccines against FAdV-4 infection remain scarce. Here, we prepared a subunit vaccine candidate derived from the bacterially expressed recombinant Fiber2 protein (termed as rFiber2 subunit vaccine) of FAdV-4 GZ-QL strain (a hypervirulent strain isolated in Guizhou province) and a recombinant plasmid pVAX1-Fiber2 as DNA vaccine candidate (termed as Fiber2 DNA vaccine). The immune effects of different dosages (50, 100, and 150 μg) of these were evaluated through immunization and challenge studies in chickens. Three injections of the rFiber2 subunit vaccine or the Fiber2 DNA vaccine induced robust humoral and cellular immune responses in chickens, which was assessed based on the secretion of high-level neutralizing antibodies, Th1- (IL-2, IFN-γ) and Th2-type cytokines (IL-4, IL-6). Importantly, the efficacy of the rFiber2 subunit vaccine was significantly higher (80 %-100 %) compared with the Fiber2 DNA vaccine (50 %-60 %) and a commercial inactivated vaccine (80 %). Collectively, these results suggest that the rFiber2 subunit and Fiber2 DNA vaccine candidate induced remarkable humoral and cellular immune responses, while the rFiber2 subunit vaccine candidate possesses better potential in the fight against FAdV-4 infection, laying foundations for the effective control of HHS in chickens.
Collapse
Affiliation(s)
- Dejing Yin
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Ling He
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Erpeng Zhu
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Tian Fang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Jun Yue
- Animal Disease Prevention and Control Center of Guizhou Province, Guiyang, 550001, China
| | - Ming Wen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Kaigong Wang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Zhentao Cheng
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
14
|
Feiz Haddad MH, Lomei J, Shokri A, Habibpour H, Rezvan H, Nourian A, Mahmoudi MR. Review of Development of Live Vaccines against Leishmaniasis. JOURNAL OF CHILD SCIENCE 2021. [DOI: 10.1055/s-0041-1731336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractLeishmaniasis is a serious public health problem in both tropical and temperate regions, caused by protozoan parasites of the genus Leishmania. Cutaneous leishmaniasis is the most common form of leishmaniasis worldwide. After recovery from the initial infection in most of the patients, a long-lasting natural immunity will be established. In individuals with HIV infection or in immune deficient patients, the more dangerous forms can occur. Despite many attempts, there is no efficient vaccine for leishmaniasis. The main concern for live-attenuated vaccines is the possibility of returning to the virulent form. Therefore, the safety is an important point in designing a successful vaccine. Nonvirulent parasites as vaccine candidates are achievable through gamma-irradiation, long-term culture, random mutations induced by chemical agents, and temperature-sensitive mutations. The type of change(s) in such parasites is not known well and drawbacks such as reversion to virulent forms was soon realized. Leishmania tarentolae with capacity of adaptation to mammalian system has a potential to be used as nonpathogenic vector in vaccine programs. Due to its nonpathogenic intrinsic property, it does not have the ability to replace with the pathogen form. Moreover, the main problems are associated with the production of live vaccines, including lyophilization, storage, standards, and quality control that must be considered. In this review, we focused on the importance of different approaches concerning the development of a live vaccine against leishmaniasis.
Collapse
Affiliation(s)
- Mohammad Hossein Feiz Haddad
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Parasitology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Jalal Lomei
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Azar Shokri
- Vector-borne Disease Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Habib Habibpour
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Parasitology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Rezvan
- Department of Pathobiology, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Alireza Nourian
- Department of Pathobiology, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Mohammad Reza Mahmoudi
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
15
|
Wu J, Abraham SN. The Roles of T cells in Bladder Pathologies. Trends Immunol 2021; 42:248-260. [PMID: 33536141 PMCID: PMC7914211 DOI: 10.1016/j.it.2021.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/26/2022]
Abstract
T lymphocytes play important roles in the skin and mucosal surfaces such as the gut and lung. Until recently the contributions of T cells to mammalian bladder immunity were largely unknown. With newer techniques, including single-cell RNA sequencing and reporter mice, an understanding is emerging of T cell roles in bladder diseases (bacterial infections, bladder cancer, chronic inflammation). In these pathologies, many bladder T cell responses can be harmful to the host through suboptimal clearance of bacteria or cancer cells, or by modulating autoinflammation. Recent findings suggest that T cell behavior might be influenced by resident T cell interactions with the bladder microbiota and other immunostimulants. Thus, regulating bladder T cell functions could emerge as a putative immunotherapy to treat some bladder diseases.
Collapse
Affiliation(s)
- Jianxuan Wu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Soman N Abraham
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore.
| |
Collapse
|
16
|
Liu L, Sun XY, Lu Y, Song JK, Xing M, Chen X, Luo Y, Ru Y, Chen ST, Li HJ, Li B, Li X. Fire Needle Therapy for the Treatment of Psoriasis: A Quantitative Evidence Synthesis. J Altern Complement Med 2021; 27:24-37. [PMID: 32757941 DOI: 10.1089/acm.2019.0409] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Liu Liu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-ying Sun
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yi Lu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian-kun Song
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Xing
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xi Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Si-ting Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong-jin Li
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Singh SP, Kumar S, Mathan SV, Tomar MS, Singh RK, Verma PK, Kumar A, Kumar S, Singh RP, Acharya A. Therapeutic application of Carica papaya leaf extract in the management of human diseases. Daru 2020; 28:735-744. [PMID: 32367410 PMCID: PMC7704890 DOI: 10.1007/s40199-020-00348-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Papaya (Carica papaya Linn.) belongs to the family Caricaceae and is well known for its therapeutic and nutritional properties all over the world. The different parts of the papaya plant have been used since ancient times for its therapeutic applications. Herein, we aimed to review the anticancer, anti-inflammatory, antidiabetic and antiviral activities of papaya leaf. METHODS All information presented in this review article regarding the therapeutic application of Carica papaya leaf extract has been acquired by approaching various electronic databases, including Scopus, Google scholar, Web of science, and PubMed. The keywords Carica papaya, anticancer, anti-inflammatory, immunomodulatory, and phytochemicals were explored until December 2019. RESULTS The papaya plant, including fruit, leaf, seed, bark, latex, and their ingredients play a major role in the management of disease progression. Carica papaya leaf contains active components such as alkaloids, glycosides, tannins, saponins, and flavonoids, which are responsible for its medicinal activity. Additionally, the leaf juice of papaya increases the platelet counts in people suffering from dengue fever. CONCLUSION The major findings revealed that papaya leaf extract has strong medicinal properties such as antibacterial, antiviral, antitumor, hypoglycaemic and anti-inflammatory activity. Furthermore, clinical trials are needed to explore the medicative potential of papaya leaf. Graphical abstract Graphical abstract showing the medicinal properties of Carica papaya leaf.
Collapse
Affiliation(s)
- Surya P Singh
- Department of Zoology, Banaras Hindu University, Varanasi, UP, India
| | - Sanjay Kumar
- Cancer and Radiation Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sivapar V Mathan
- Cancer and Radiation Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Rishi Kant Singh
- Department of Zoology, Banaras Hindu University, Varanasi, UP, India
| | | | - Amit Kumar
- Department of Zoology, Banaras Hindu University, Varanasi, UP, India
| | - Sandeep Kumar
- Department of Zoology, Banaras Hindu University, Varanasi, UP, India
| | - Rana P Singh
- Cancer and Radiation Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.
| | - Arbind Acharya
- Department of Zoology, Banaras Hindu University, Varanasi, UP, India.
| |
Collapse
|
18
|
Andrianov AK, Marin A, Wang R, Karauzum H, Chowdhury A, Agnihotri P, Yunus AS, Mariuzza RA, Fuerst TR. Supramolecular assembly of Toll-like receptor 7/8 agonist into multimeric water-soluble constructs enables superior immune stimulation in vitro and in vivo. ACS APPLIED BIO MATERIALS 2020; 3:3187-3195. [PMID: 33880435 DOI: 10.1021/acsabm.0c00189] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Resiquimod or R848 (RSQD) is a Toll-like receptor (TLR) 7/8 agonist which shows promise as vaccine adjuvant due to its potential to promote highly desirable cellular immunity. The development of this small molecule in the field to date has been largely impeded by its rapid in vivo clearance and lack of association with vaccine antigens. Here, we report a multimeric TLR 7/8 construct of nano-scale size, which results from a spontaneous self-assembly of RSQD with a water-soluble clinical-stage polymer - poly[di(carboxylatophenoxy)phosphazene] (PCPP). The formation of ionically paired construct (PCPP-R) and a ternary complex, which also includes Hepatitis C virus (HCV) antigen, has been demonstrated by dynamic lights scattering (DLS), turbidimetry, fluorescence spectroscopy, asymmetric flow field flow fractionation (AF4), and 1H NMR spectroscopy methods. The resulting supramolecular assembly PCPP-R enabled superior immunostimulation in cellular assays (mouse macrophage reporter cell line) and displayed improved in vitro hemocompatibility (human erythrocytes). In vivo studies demonstrated that PCPP-R adjuvanted HCV formulation induced higher serum neutralization titers in BALB/c mice and shifted the response towards desirable cellular immunity, as evaluated by antibody isotype ratio (IgG2a/IgG1) and ex vivo analysis of cytokine secreting splenocytes (higher levels of interferon gamma (IFN-γ) single and tumor necrosis factor alpha (TNF-α)/IFN-γ double producing cells). The non-covalent multimerization approach stands in contrast to previously suggested RSQD delivery methods, which involve covalent conjugation or encapsulation, and offers a flexible methodology that can be potentially integrated with other parenterally administered drugs.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850, USA
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850, USA
| | - Ruixue Wang
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850, USA
| | | | - Ananda Chowdhury
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850, USA
| | - Pragati Agnihotri
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, USA.,W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850
| | - Abdul S Yunus
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850, USA
| | - Roy A Mariuzza
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850, USA.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, USA.,W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850, USA.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, USA
| |
Collapse
|
19
|
Pagheh AS, Sarvi S, Gholami S, Asgarian-Omran H, Valadan R, Hassannia H, Ahmadpour E, Fasihi-Ramandie M, Dodangeh S, Hosseni-khah Z, Daryani A. Protective efficacy induced by DNA prime and recombinant protein boost vaccination with Toxoplasma gondii GRA14 in mice. Microb Pathog 2019; 134:103601. [DOI: 10.1016/j.micpath.2019.103601] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
|
20
|
Association between Inflammatory Cytokine Levels and Thrombocytopenia during Plasmodium falciparum and P. vivax Infections in South-Western Coastal Region of India. Malar Res Treat 2019; 2019:4296523. [PMID: 31110658 PMCID: PMC6487116 DOI: 10.1155/2019/4296523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/11/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Background Thrombocytopenia is a most commonly observed complication during malaria infections. Inflammatory cytokines such as IL-1, IL-6, and IL-10 have been documented in malaria induced thrombocytopaenia. This study was aimed to understand the possible relationship between inflammatory cytokines across varying degrees of thrombocytopenia during P. vivax, P. falciparum, and mixed infections. Methods A hospital-based cross sectional study was conducted at District Wenlock Hospital in Mangaluru, a city situated along the south-western coastal region of Arabian Sea in India. In this study, blood samples from 627 malaria patients were analyzed for infected parasite species, clinical conditions, platelet levels, and key cytokines that are produced in response to infection; samples from 176 uninfected healthy individuals were used as controls. Results The results of our study showed a high prevalence of malarial thrombocytopenia (platelets <150 ×103/μl) in this endemic settings. About 62.7% patients had mild-to-moderate levels of thrombocytopenia and 16% patients had severe thrombocytopenia (platelets <50 × 103/μl). Upon comparison of cytokines across varying degrees of thrombocytopenia, irrespective of infecting species, the levels of TNF-α and IL-10 were significantly higher during thrombocytopenia, whereas IL-6 levels were considerably lower in severe thrombocytopenia patients suffering from P. vivax or P. falciparum infections. The severe clinical complications observed in patients with malarial thrombocytopenia included severe anemia (17.5%), acute renal failure (12.7%), jaundice (27.0%), metabolic acidosis (36.5%), spontaneous bleeding (3.2%), hypoglycemia (25.4%), hyperparasitemia (4.8%), acute respiratory distress syndrome (1.6%), pulmonary edema (19.0%), and cerebral malaria (1.6%) in various combinations. Conclusion Overall, the results of our study suggest that inflammatory cytokines influence the transformation of mild forms of thrombocytopenia into severe forms during malarial infections. Further studies are needed to understand the association of inflammatory cytokine responses with severe malaria complications and thrombocytopenia.
Collapse
|
21
|
Goldberg MF, Roeske EK, Ward LN, Pengo T, Dileepan T, Kotov DI, Jenkins MK. Salmonella Persist in Activated Macrophages in T Cell-Sparse Granulomas but Are Contained by Surrounding CXCR3 Ligand-Positioned Th1 Cells. Immunity 2018; 49:1090-1102.e7. [PMID: 30552021 DOI: 10.1016/j.immuni.2018.10.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/16/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
Abstract
Salmonella enterica (Se) bacteria cause persistent intracellular infections while stimulating a robust interferon-γ-producing CD4+ T (Th1) cell response. We addressed this paradox of concomitant infection and immunity by tracking fluorescent Se organisms in mice. Se bacteria persisted in nitric oxide synthase (iNOS)-producing resident and recruited macrophages while inducing genes related to protection from nitric oxide. Se-infected cells occupied iNOS+ splenic granulomas that excluded T cells but were surrounded by mononuclear phagocytes producing the chemokines CXCL9 and CXCL10, and Se epitope-specific Th1 cells expressing CXCR3, the receptor for these chemokines. Blockade of CXCR3 inhibited Th1 occupancy of CXCL9/10-dense regions, reduced activation of the Th1 cells, and led to increased Se growth. Thus, intracellular Se bacteria survive in their hosts by counteracting toxic products of the innate immune response and by residing in T cell-sparse granulomas, away from abundant Th1 cells positioned via CXCR3 in a bordering region that act to limit infection.
Collapse
Affiliation(s)
- Michael F Goldberg
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Elizabeth K Roeske
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Lauren N Ward
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Thomas Pengo
- University of Minnesota Informatics Institute, Office of the Vice President for Research, Minneapolis, MN 55455, USA
| | - Thamotharampillai Dileepan
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Dmitri I Kotov
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Marc K Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
22
|
Growth arrested live-attenuated Leishmania infantum KHARON1 null mutants display cytokinesis defect and protective immunity in mice. Sci Rep 2018; 8:11627. [PMID: 30072701 PMCID: PMC6072785 DOI: 10.1038/s41598-018-30076-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/24/2018] [Indexed: 01/30/2023] Open
Abstract
There is no safe and efficacious vaccine against human leishmaniasis available and live attenuated vaccines have been used as a prophylactic alternative against the disease. In order to obtain an attenuated Leishmania parasite for vaccine purposes, we generated L. infantum KHARON1 (KH1) null mutants (ΔLikh1). This gene was previously associated with growth defects in L. mexicana. ΔLikh1 was obtained and confirmed by PCR, qPCR and Southern blot. We also generate a KH1 complemented line with the introduction of episomal copies of KH1. Although ΔLikh1 promastigote forms exhibited a growth pattern similar to the wild-type line, they differ in morphology without affecting parasite viability. L. infantum KH1-deficient amastigotes were unable to sustain experimental infection in macrophages, forming multinucleate cells which was confirmed by in vivo attenuation phenotype. The cell cycle analysis of ΔLikh1 amastigotes showed arrested cells at G2/M phase. ΔLikh1-immunized mice presented reduced parasite burden upon challenging with virulent L. infantum, when compared to naïve mice. An effect associated with increased Li SLA-specific IgG serum levels and IL-17 production. Thus, ΔLikh1 parasites present an infective-attenuated phenotype due to a cytokinesis defect, whereas it induces immunity against visceral leishmaniasis in mouse model, being a candidate for antileishmanial vaccine purposes.
Collapse
|
23
|
Flaxman A, Ewer KJ. Methods for Measuring T-Cell Memory to Vaccination: From Mouse to Man. Vaccines (Basel) 2018; 6:E43. [PMID: 30037078 PMCID: PMC6161152 DOI: 10.3390/vaccines6030043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022] Open
Abstract
The development of effective vaccines continues to be a key goal for public health bodies, governments, funding bodies and pharmaceutical companies. With new vaccines such as Shingrix targeting Shingles and Bexsero for Meningitis B, licensed in recent years, today's population can be protected from more infectious diseases than ever before. Despite this, we are yet to license vaccines for some of the deadliest endemic diseases affecting children, such as malaria. In addition, the threat of epidemics caused by emerging pathogens is very real as exemplified by the 2014⁻2016 Ebola outbreak. Most licensed vaccines provide efficacy through humoral immunity and correlates of protection often quantify neutralising antibody titre. The role of T-cells in vaccine efficacy is less well understood and more complex to quantify. Defining T-cell responses which afford protection also remains a challenge, although more sophisticated assays for assessing cell-mediated immunity with the potential for higher throughput and scalability are now available and warrant review. Here we discuss the benefits of multiparameter cytokine analysis and omics approaches compared with flow cytometric and ELISpot assays. We also review technical challenges unique to clinical trial studies, including assay validation across laboratories and availability of sample type. Measuring T-cell immunogenicity alongside humoral responses provides information on the breadth of immune responses induced by vaccination. Accurately enumerating and phenotyping T-cell immunogenicity to vaccination is key for the determination of immune correlates of protection. However, identifying such T-cell parameters remains challenging without a clear understanding of the immunological mechanisms by which a T-cell-mediated response induces protection.
Collapse
Affiliation(s)
- Amy Flaxman
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
24
|
Fisher DG, Coppock GM, López CB. Virus-derived immunostimulatory RNA induces type I IFN-dependent antibodies and T-cell responses during vaccination. Vaccine 2018; 36:4039-4045. [PMID: 29861183 PMCID: PMC6265594 DOI: 10.1016/j.vaccine.2018.05.100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 05/17/2018] [Accepted: 05/25/2018] [Indexed: 01/04/2023]
Abstract
Adjuvants potentiate and direct the type of immunity elicited during vaccination. However, there is a shortage of adjuvants that elicit robust type-1 immunity required for the control of intracellular pathogens, including viruses. RNA derived from Sendai virus defective viral genomes (DVGs) stimulates RIG-I-like receptor signaling leading to type-1 immunity during infection. Here, we investigated whether a 268nt DVG-derived oligonucleotide (DDO) functions as a strong type-1 immunity-inducing adjuvant during vaccination against influenza virus. We show that DDO induces robust IgG2c antibody production when used in an inactivated influenza A virus (IAV) vaccine. Additionally, DDO induces Th1 and CD8+ T-cell responses able to protect against heterosubtypic IAV challenge. Interestingly, DDO synergized with AddaVax and skewed the immune response towards type-1 immunity. The adjuvancy of DDO alone and in synergy with AddaVax was heavily dependent on type I interferon signaling. Our data support a critical role for type I interferon in the induction of type-1 immune responses during vaccination and demonstrate that DDO is a type-1 immunity orienting vaccine adjuvant that can be used alone or in synergy with other adjuvants.
Collapse
Affiliation(s)
- Devin G Fisher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Gaia M Coppock
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Carolina B López
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
25
|
Climent N, García I, Marradi M, Chiodo F, Miralles L, Maleno MJ, Gatell JM, García F, Penadés S, Plana M. Loading dendritic cells with gold nanoparticles (GNPs) bearing HIV-peptides and mannosides enhance HIV-specific T cell responses. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:339-351. [PMID: 29157976 DOI: 10.1016/j.nano.2017.11.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/05/2017] [Accepted: 11/03/2017] [Indexed: 01/10/2023]
Abstract
Gold nanoparticles (GNPs) decorated with glycans ameliorate dendritic cells (DC) uptake, antigen-presentation and T-cells cross-talk, which are important aspects in vaccine design. GNPs allow for high antigen loading, DC targeting, lack of toxicity and are straightforward prepared and easy to handle. The present study aimed to assess the capacity of DC to process and present HIV-1-peptides loaded onto GNPs bearing high-mannoside-type oligosaccharides (P1@HM) to autologous T-cells from HIV-1 patients. The results showed that P1@HM increased HIV-specific CD4+ and CD8+ T-cell proliferation and induced highly functional cytokine secretion compared with HIV-peptides alone. P1@HM elicits a highly efficient secretion of pro-TH1 cytokines and chemokines, a moderate production of pro-TH2 and significant higher secretion of pro-inflammatory cytokines such as TNF-α and IL-1β. Thus, co-delivery of HIV-1 antigens and HM by GNPs is an excellent vaccine delivery system inducing HIV-specific cellular immune responses in HIV+ patients, being a promising approach to improve anti-HIV-1 vaccines.
Collapse
Affiliation(s)
- Núria Climent
- AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), HIV Vaccine Development in Catalonia (HIVACAT), Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Isabel García
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Paseo Miramón 182, Donostia-San Sebastián, Spain; CIC biomaGUNE, Paseo de Miramón 182, Donostia-San Sebastián, Spain
| | - Marco Marradi
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Paseo Miramón 182, Donostia-San Sebastián, Spain; CIC biomaGUNE, Paseo de Miramón 182, Donostia-San Sebastián, Spain
| | - Fabrizio Chiodo
- CIC biomaGUNE, Paseo de Miramón 182, Donostia-San Sebastián, Spain; Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, The Netherland
| | - Laia Miralles
- AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), HIV Vaccine Development in Catalonia (HIVACAT), Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - María José Maleno
- AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), HIV Vaccine Development in Catalonia (HIVACAT), Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - José María Gatell
- AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), HIV Vaccine Development in Catalonia (HIVACAT), Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain; Service of Infectious Diseases & AIDS Unit, Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Felipe García
- AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), HIV Vaccine Development in Catalonia (HIVACAT), Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain; Service of Infectious Diseases & AIDS Unit, Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Soledad Penadés
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Paseo Miramón 182, Donostia-San Sebastián, Spain; CIC biomaGUNE, Paseo de Miramón 182, Donostia-San Sebastián, Spain
| | - Montserrat Plana
- AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), HIV Vaccine Development in Catalonia (HIVACAT), Hospital Clínic de Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
26
|
Ford T, Wenden C, Mbekeani A, Dally L, Cox JH, Morin M, Winstone N, Hill AVS, Gilmour J, Ewer KJ. Cryopreservation-related loss of antigen-specific IFNγ producing CD4 + T-cells can skew immunogenicity data in vaccine trials: Lessons from a malaria vaccine trial substudy. Vaccine 2017; 35:1898-1906. [PMID: 28285985 PMCID: PMC5387668 DOI: 10.1016/j.vaccine.2017.02.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/31/2017] [Accepted: 02/18/2017] [Indexed: 12/19/2022]
Abstract
Ex vivo functional immunoassays such as ELISpot and intracellular cytokine staining (ICS) by flow cytometry are crucial tools in vaccine development both in the identification of novel immunogenic targets and in the immunological assessment of samples from clinical trials. Cryopreservation and subsequent thawing of PBMCs via validated processes has become a mainstay of clinical trials due to processing restrictions inherent in the disparate location and capacity of trial centres, and also in the need to standardize biological assays at central testing facilities. Logistical and financial requirement to batch process samples from multiple study timepoints are also key. We used ELISpot and ICS assays to assess antigen-specific immunogenicity in blood samples taken from subjects enrolled in a phase II malaria heterologous prime-boost vaccine trial and showed that the freeze thaw process can result in a 3–5-fold reduction of malaria antigen-specific IFNγ-producing CD3+CD4+ effector populations from PBMC samples taken post vaccination. We have also demonstrated that peptide responsive CD8+ T cells are relatively unaffected, as well as CD4+ T cell populations that do not produce IFNγ. These findings contribute to a growing body of data that could be consolidated and synthesised as guidelines for clinical trials with the aim of increasing the efficiency of vaccine development pipelines.
Collapse
Affiliation(s)
- Tom Ford
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK.
| | - Claire Wenden
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Alison Mbekeani
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Len Dally
- EMMES Corporation, Rockville, MD, USA
| | - Josephine H Cox
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK
| | | | - Nicola Winstone
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Adrian V S Hill
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford, UK
| | - Jill Gilmour
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Katie J Ewer
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford, UK
| |
Collapse
|
27
|
Francica JR, Lynn GM, Laga R, Joyce MG, Ruckwardt TJ, Morabito KM, Chen M, Chaudhuri R, Zhang B, Sastry M, Druz A, Ko K, Choe M, Pechar M, Georgiev IS, Kueltzo LA, Seymour LW, Mascola JR, Kwong PD, Graham BS, Seder RA. Thermoresponsive Polymer Nanoparticles Co-deliver RSV F Trimers with a TLR-7/8 Adjuvant. Bioconjug Chem 2016; 27:2372-2385. [PMID: 27583777 DOI: 10.1021/acs.bioconjchem.6b00370] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Structure-based vaccine design has been used to develop immunogens that display conserved neutralization sites on pathogens such as HIV-1, respiratory syncytial virus (RSV), and influenza. Improving the immunogenicity of these designed immunogens with adjuvants will require formulations that do not alter protein antigenicity. Here, we show that nanoparticle-forming thermoresponsive polymers (TRP) allow for co-delivery of RSV fusion (F) protein trimers with Toll-like receptor 7 and 8 agonists (TLR-7/8a) to enhance protective immunity. Although primary amine conjugation of TLR-7/8a to F trimers severely disrupted the recognition of critical neutralizing epitopes, F trimers site-selectively coupled to TRP nanoparticles retained appropriate antigenicity and elicited high titers of prefusion-specific, TH1 isotype anti-RSV F antibodies following vaccination. Moreover, coupling F trimers to TRP delivering TLR-7/8a resulted in ∼3-fold higher binding and neutralizing antibody titers than soluble F trimers admixed with TLR-7/8a and conferred protection from intranasal RSV challenge. Overall, these data show that TRP nanoparticles may provide a broadly applicable platform for eliciting neutralizing antibodies to structure-dependent epitopes on RSV, influenza, HIV-1, or other pathogens.
Collapse
Affiliation(s)
- Joseph R Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Geoffrey M Lynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Richard Laga
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic , 162 06 Prague, Czech Republic
| | - M Gordon Joyce
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Kaitlyn M Morabito
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Man Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Rajoshi Chaudhuri
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Gaithersburg, Maryland 20878, United States
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Mallika Sastry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Aliaksandr Druz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Kiyoon Ko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Misook Choe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic , 162 06 Prague, Czech Republic
| | - Ivelin S Georgiev
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Lisa A Kueltzo
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Gaithersburg, Maryland 20878, United States
| | | | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| |
Collapse
|
28
|
Bautista BL, Devarajan P, McKinstry KK, Strutt TM, Vong AM, Jones MC, Kuang Y, Mott D, Swain SL. Short-Lived Antigen Recognition but Not Viral Infection at a Defined Checkpoint Programs Effector CD4 T Cells To Become Protective Memory. THE JOURNAL OF IMMUNOLOGY 2016; 197:3936-3949. [PMID: 27798159 DOI: 10.4049/jimmunol.1600838] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/19/2016] [Indexed: 01/20/2023]
Abstract
Although memory CD4 T cells are critical for effective immunity to pathogens, the mechanisms underlying their generation are still poorly defined. We find that following murine influenza infection, most effector CD4 T cells undergo apoptosis unless they encounter cognate Ag at a defined stage near the peak of effector generation. Ag recognition at this memory checkpoint blocks default apoptosis and programs their transition to long-lived memory. Strikingly, we find that viral infection is not required, because memory formation can be restored by the addition of short-lived, Ag-pulsed APC at this checkpoint. The resulting memory CD4 T cells express an enhanced memory phenotype, have increased cytokine production, and provide protection against lethal influenza infection. Finally, we find that memory CD4 T cell formation following cold-adapted influenza vaccination is boosted when Ag is administered during this checkpoint. These findings imply that persistence of viral Ag presentation into the effector phase is the key factor that determines the efficiency of memory generation. We also suggest that administering Ag at this checkpoint may improve vaccine efficacy.
Collapse
Affiliation(s)
- Bianca L Bautista
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | | | - K Kai McKinstry
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Tara M Strutt
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Allen M Vong
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Michael C Jones
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Yi Kuang
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Daniel Mott
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605
| |
Collapse
|
29
|
Lin X, Xiao G, Luo D, Kong L, Chen X, Sun D, Yan J. Chimeric epitope vaccine against Leptospira interrogans infection and induced specific immunity in guinea pigs. BMC Microbiol 2016; 16:241. [PMID: 27737644 PMCID: PMC5064800 DOI: 10.1186/s12866-016-0852-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2016] [Indexed: 11/28/2022] Open
Abstract
Background Leptospirosis is an important reemerging zoonosis, with more than half a million cases reported annually, and is caused by pathogenic Leptospira species. Development of a universal vaccine is one of the major strategic goals to overcome the disease burden of leptospirosis. In this study, a chimeric multi-epitope protein-based vaccine was designed and tested for its potency to induce a specific immune response and provide protection against L. interrogans infection. Results The protein, containing four repeats of six T- and B-cell combined epitopes from the leptospiral outer membrane proteins, OmpL1, LipL32 and LipL21, was expressed and purified. Western blot analysis showed that the recombinant protein (named r4R) mainly expressed in a soluble pattern, and reacted with antibodies raised in rabbit against heat-killed Leptospira and in guinea pigs against the r4R vaccine. Microscopic agglutination tests showed that r4R antisera was immunological cross-reactive with a range of Chinese standard reference strains of Leptospira belonging to different serogroups. In guinea pigs, the r4R vaccine induced a Th1-biased immune response, as reflected by the IgG2a/IgG1 ratio and cytokine production of stimulated splenocytes derived from immunized animals. Finally, r4R-immunized guinea pigs showed increased survival of lethal Leptospira challenges compared with PBS-immunized animals and tissue damage and leptospiral colonization of the kidney were reduced. Conclusions The multi-epitope chimeric r4R protein is a promising antigen for the development of a universal cross-reactive vaccine against leptospirosis. Electronic supplementary material The online version of this article (doi:10.1186/s12866-016-0852-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xu'ai Lin
- Department of Medical Microbiology and Parasitology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China. .,Basic Medical Microbiology Division, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| | - Guohui Xiao
- Department of Medical Microbiology and Parasitology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Dongjiao Luo
- Department of Medical Microbiology and Parasitology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Liangliang Kong
- Department of Medical Microbiology and Parasitology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Xu Chen
- Department of Medical Microbiology and Parasitology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Dexter Sun
- Department of Neurology and Neuroscience, New York Presbyterian Hospital and Hospital for Special Surgery, Cornell University Weill Medical College, New York, NY, 10021, USA
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China. .,Basic Medical Microbiology Division, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
30
|
da Cunha A, Antoniazi Michelin M, Cândido Murta EF. Phenotypic profile of dendritic and T cells in the lymph node of Balb/C mice with breast cancer submitted to dendritic cells immunotherapy. Immunol Lett 2016; 177:25-37. [PMID: 27423825 DOI: 10.1016/j.imlet.2016.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/01/2016] [Accepted: 07/12/2016] [Indexed: 11/25/2022]
Abstract
Breast cancer (BC) is the most common malignant neoplasm and the cause of death by cancer among women worldwide. Its development influenced by various mutations that occur in the tumor cell and by the immune system's status, which has a direct influence on the tumor microenvironment and, consequently, on interactions with non-tumor cells involved in the immunological response. Strategies using dendritic cells (DCs) or antigen-presenting cells (APCs), therapeutic mode, in cancer have been developed for some time. The proper interaction between DCs and T cells upon antigen presentation is of greatest importance for an antitumor immune response activation. Thus, various receptors on the surface of T cells must be able to recognize ligands that are located on the surface of APCs. However, little is known about the real behavior and interaction forms of CDs and T cells after vaccination. Due to the crucial importance of DCs in an effective anti-tumor immune response activation and the search for compliant results in inducing this response by immunotherapies with DCs, the phenotypic profile of DCs and T cells in lymph nodes obtained from female Balb/C mice with breast cancer induced by 4T1 cells and DCs treated with vaccines was investigated. We evaluated through flow cytometry based on the surface and intracellular molecules marking; as well as the presence of cytokines and chemokines, IL-2, IL-4, IL-10, IL-12, IFN-γ, TNF-α and TGF-β in the supernatant of the culture of Balb/C lymph nodes by ELISA. The results show that the vaccination with DCs, in the maturation parameters used in this study, was able to stimulate the secretion of cytokines such as IFN-γ and IL-12 and inhibit the secretion of TGF-β and IL-10 in nodal lymph infiltrates, as well as co-stimulatory activating (CD86) and adhesion molecules in DCs and T cells LFA-1/ICAM-1 and inhibit the secretion of CTLA-4 present in lymph nodes. Facts that led to aTh1 profile polarization, immuno competent in relation to breast cancer. We indirectly evaluated the interaction between DCs and T cells dependent on the vaccination with DCs in tumor draining lymph nodes, in breast cancer in Balb/C mice and we believe that, this way, we will be able to achieve a model vaccine protocol in the future, based on the correct interaction between cells that enable the induction of anti-tumor effective response. Breast cancer (BC) is the most common malignant neoplasm and the cause of death by cancer among women worldwide. Its development influenced by various mutations that occur in the tumor cell and by the immune system's status, which has a direct influence on the tumor microenvironment and, consequently, on interactions with non-tumor cells involved in the immunological response. Strategies using dendritic cells (DCs) or antigen-presenting cells (APCs), therapeutic mode, in cancer have been developed for some time. The proper interaction between DCs and T cells upon antigen presentation is of greatest importance for an antitumor immune response activation. Thus, various receptors on the surface of T cells must be able to recognize ligands that are located on the surface of APCs. However, little is known about the real behavior and interaction forms of DCs and T cells after vaccination. Due to the crucial importance of DCs in an effective anti-tumor immune response activation and the search for compliant results in inducing this response by immunotherapies with DCs, the phenotypic profile of DCs and T cells in lymph nodes obtained from female Balb/C mice with breast cancer induced by 4T1 cells and DCs treated with vaccines was investigated. We evaluated through flow cytometry based on the surface and intracellular molecules marking; as well as the presence of cytokines and chemokines, IL-2, IL-4, IL-10, IL-12, IFN-γ, TNF-α and TGF-β in the supernatant of the culture of Balb/C lymph nodes by ELISA. The results show that the vaccination with DCs, in the maturation parameters used in this study, was able to stimulate the secretion of cytokines such as IFN-γ and IL-12 and inhibit the secretion of TGF-β and IL-10 in nodal lymph infiltrates, as well as co-stimulatory activating (CD86) and adhesion molecules in DCs and T cells LFA-1/ICAM-1 and inhibit the secretion of CTLA-4 present in lymph nodes. Facts that led to aTh1 profile polarization, immuno competent in relation to breast cancer. We indirectly evaluated the interaction between DCs and T cells dependent on the vaccination with DCs in tumor draining lymph nodes, in breast cancer in Balb/C mice and we believe that, this way, we will be able to achieve a model vaccine protocol in the future, based on the correct interaction between cells that enable the induction of anti-tumor effective response.
Collapse
Affiliation(s)
- Alessandra da Cunha
- Research Oncology Institute (IPON), Federal University of the Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-440, Brazil
| | - Marcia Antoniazi Michelin
- Discipline of Immunology, Federal University of the Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-440, Brazil
| | - Eddie Fernando Cândido Murta
- Discipline of Gynecology and Obstetrics, Federal University of the Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, 38025-440, Brazil.
| |
Collapse
|
31
|
Liu SD, Su J, Zhang SM, Dong HP, Wang H, Luo W, Wen Q, He JC, Yang XF, Ma L. Identification of HLA-A*11:01-restricted Mycobacterium tuberculosis CD8(+) T cell epitopes. J Cell Mol Med 2016; 20:1718-28. [PMID: 27072810 PMCID: PMC4988290 DOI: 10.1111/jcmm.12867] [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: 10/28/2015] [Accepted: 03/07/2016] [Indexed: 01/06/2023] Open
Abstract
New vaccines are needed to combat Mycobacterium tuberculosis (MTB) infections. The currently employed Bacillus Calmette‐Guérin vaccine is becoming ineffective, due in part to the emergence of multidrug‐resistant tuberculosis (MDR‐TB) strains and the reduced immune capacity in cases of HIV coinfection. CD8+ T cells play an important role in the protective immunity against MTB infections, and the identification of immunogenic CD8+ T cell epitopes specific for MTB is essential for the design of peptide‐based vaccines. To identify CD8+ T cell epitopes of MTB proteins, we screened a set of 94 MTB antigens for HLA class I A*11:01‐binding motifs. HLA‐A*11:01 is one of the most prevalent HLA molecules in Southeast Asians, and definition of T cell epitopes it can restrict would provide significant coverage for the Asian population. Peptides that bound with high affinity to purified HLA molecules were subsequently evaluated in functional assays to detect interferon‐γ release and CD8+ T cell proliferation in active pulmonary TB patients. We identified six novel epitopes, each derived from a unique MTB antigen, which were recognized by CD8+ T cells from active pulmonary TB patients. In addition, a significant level of epitope‐specific T cells could be detected ex vivo in peripheral blood mononuclear cells from active TB patients by an HLA‐A*11:01 dextramer carrying the peptide Rv3130c194‐204 (from the MTB triacylglycerol synthase Tgs1), which was the most frequently recognized epitope in our peptide library. In conclusion, this study identified six dominant CD8+ T cell epitopes that may be considered potential targets for subunit vaccines or diagnostic strategies against TB.
Collapse
Affiliation(s)
- Su-Dong Liu
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Jin Su
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shi-Meng Zhang
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Hai-Ping Dong
- Department of Severe Tuberculosis Medicine, Guangzhou Chest Hospital, Guangzhou, China
| | - Hui Wang
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Wei Luo
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Qian Wen
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Jian-Chun He
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiao-Fan Yang
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Li Ma
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
32
|
Aberle JH, Schwaiger J, Aberle SW, Stiasny K, Scheinost O, Kundi M, Chmelik V, Heinz FX. Human CD4+ T Helper Cell Responses after Tick-Borne Encephalitis Vaccination and Infection. PLoS One 2015; 10:e0140545. [PMID: 26465323 PMCID: PMC4605778 DOI: 10.1371/journal.pone.0140545] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/28/2015] [Indexed: 12/30/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) is a human-pathogenic flavivirus that is endemic in large parts of Europe and Asia and causes severe neuroinvasive illness. A formalin-inactivated vaccine induces strong neutralizing antibody responses and confers protection from TBE disease. CD4+ T cell responses are essential for neutralizing antibody production, but data on the functionalities of TBEV-specific CD4+ T cells in response to vaccination or infection are lacking. This study provides a comprehensive analysis of the cytokine patterns of CD4+ T cell responses in 20 humans after TBE vaccination in comparison to those in 18 patients with TBEV infection. Specifically, Th1-specific cytokines (IFN-γ, IL-2, TNF-α), CD40 ligand and the Th1 lineage-specifying transcription factor Tbet were determined upon stimulation with peptides covering the TBEV structural proteins contained in the vaccine (C-capsid, prM/M-membrane and E-envelope). We show that TBEV-specific CD4+ T cell responses are polyfunctional, but the cytokine patterns after vaccination differed from those after infection. TBE vaccine responses were characterized by lower IFN-γ responses and high proportions of TNF-α+IL-2+ cells. In vaccine-induced responses—consistent with the reduced IFN-γ expression patterns—less than 50% of TBEV peptides were detected by IFN-γ+ cells as compared to 96% detected by IL-2+ cells, indicating that the single use of IFN-γ as a read-out strongly underestimates the magnitude and breadth of such responses. The results provide important insights into the functionalities of CD4+ T cells that coordinate vaccine responses and have direct implications for future studies that address epitope specificity and breadth of these responses.
Collapse
Affiliation(s)
- Judith H. Aberle
- Department of Virology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| | - Julia Schwaiger
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Stephan W. Aberle
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Ondrej Scheinost
- Laboratory of Molecular Genetics, Hospital České Budĕjovice, České Budĕjovice, Czech Republic
| | - Michael Kundi
- Centre for Public Health, Medical University of Vienna, Vienna, Austria
| | - Vaclav Chmelik
- Department of Infectious Diseases, Hospital České Budĕjovice, České Budĕjovice, Czech Republic
| | - Franz X. Heinz
- Department of Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
33
|
Beljanski V, Chiang C, Kirchenbaum GA, Olagnier D, Bloom CE, Wong T, Haddad EK, Trautmann L, Ross TM, Hiscott J. Enhanced Influenza Virus-Like Particle Vaccination with a Structurally Optimized RIG-I Agonist as Adjuvant. J Virol 2015; 89:10612-24. [PMID: 26269188 PMCID: PMC4580177 DOI: 10.1128/jvi.01526-15] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The molecular interaction between viral RNA and the cytosolic sensor RIG-I represents the initial trigger in the development of an effective immune response against infection with RNA viruses, resulting in innate immune activation and subsequent induction of adaptive responses. In the present study, the adjuvant properties of a sequence-optimized 5'-triphosphate-containing RNA (5'pppRNA) RIG-I agonist (termed M8) were examined in combination with influenza virus-like particles (VLP) (M8-VLP) expressing H5N1 influenza virus hemagglutinin (HA) and neuraminidase (NA) as immunogens. In combination with VLP, M8 increased the antibody response to VLP immunization, provided VLP antigen sparing, and protected mice from a lethal challenge with H5N1 influenza virus. M8-VLP immunization also led to long-term protective responses against influenza virus infection in mice. M8 adjuvantation of VLP increased endpoint and antibody titers and inhibited influenza virus replication in lungs compared with approved or experimental adjuvants alum, AddaVax, and poly(I·C). Uniquely, immunization with M8-VLP stimulated a TH1-biased CD4 T cell response, as determined by increased TH1 cytokine levels in CD4 T cells and increased IgG2 levels in sera. Collectively, these data demonstrate that a sequence-optimized, RIG-I-specific agonist is a potent adjuvant that can be utilized to increase the efficacy of influenza VLP vaccination and dramatically improve humoral and cellular mediated protective responses against influenza virus challenge. IMPORTANCE The development of novel adjuvants to increase vaccine immunogenicity is an important goal that seeks to improve vaccine efficacy and ultimately prevent infections that endanger human health. This proof-of-principle study investigated the adjuvant properties of a sequence-optimized 5'pppRNA agonist (M8) with enhanced capacity to stimulate antiviral and inflammatory gene networks using influenza virus-like particles (VLP) expressing HA and NA as immunogens. Vaccination with VLP in combination with M8 increased anti-influenza virus antibody titers and protected animals from lethal influenza virus challenge, highlighting the potential clinical use of M8 as an adjuvant in vaccine development. Altogether, the results describe a novel immunostimulatory agonist targeted to the cytosolic RIG-I sensor as an attractive vaccine adjuvant candidate that can be used to increase vaccine efficacy, a pressing issue in children and the elderly population.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Animals
- Antibodies, Viral/biosynthesis
- DEAD Box Protein 58
- DEAD-box RNA Helicases/chemistry
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/immunology
- Dendritic Cells/immunology
- Dendritic Cells/virology
- Female
- HEK293 Cells
- Hemagglutinins, Viral/chemistry
- Hemagglutinins, Viral/genetics
- Hemagglutinins, Viral/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunization
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice
- Mice, Inbred BALB C
- Neuraminidase/chemistry
- Neuraminidase/genetics
- Neuraminidase/immunology
- Oligoribonucleotides/administration & dosage
- Oligoribonucleotides/genetics
- Oligoribonucleotides/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/mortality
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Primary Cell Culture
- Receptors, Immunologic
- Survival Analysis
- Th1-Th2 Balance/drug effects
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Vladimir Beljanski
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Cindy Chiang
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Greg A Kirchenbaum
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - David Olagnier
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Chalise E Bloom
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Terianne Wong
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Elias K Haddad
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Lydie Trautmann
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Ted M Ross
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - John Hiscott
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| |
Collapse
|
34
|
Naval-Macabuhay I, Casanova V, Navarro G, García F, León A, Miralles L, Rovira C, Martinez-Navio JM, Gallart T, Mallol J, Gatell JM, Lluís C, Franco R, McCormick PJ, Climent N. Adenosine deaminase regulates Treg expression in autologous T cell-dendritic cell cocultures from patients infected with HIV-1. J Leukoc Biol 2015; 99:349-59. [PMID: 26310829 DOI: 10.1189/jlb.3a1214-580rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 08/15/2015] [Indexed: 12/26/2022] Open
Abstract
Regulatory T cells have an important role in immune suppression during HIV-1 infection. As regulatory T cells produce the immunomodulatory molecule adenosine, our aim here was to assess the potential of adenosine removal to revert the suppression of anti-HIV responses exerted by regulatory T cells. The experimental setup consisted of ex vivo cocultures of T and dendritic cells, to which adenosine deaminase, an enzyme that hydrolyzes adenosine, was added. In cells from healthy individuals, adenosine hydrolysis decreased CD4(+)CD25(hi) regulatory T cells. Addition of 5'-N-ethylcarboxamidoadenosine, an adenosine receptor agonist, significantly decreased CD4(+)CD25(lo) cells, confirming a modulatory role of adenosine acting via adenosine receptors. In autologous cocultures of T cells with HIV-1-pulsed dendritic cells, addition of adenosine deaminase led to a significant decrease of HIV-1-induced CD4(+)CD25(hi) forkhead box p3(+) cells and to a significant enhancement of the HIV-1-specific CD4(+) responder T cells. An increase in the effector response was confirmed by the enhanced production of CD4(+) and CD8(+) CD25(-)CD45RO(+) memory cell generation and secretion of Th1 cytokines, including IFN-γ and IL-15 and chemokines MIP-1α/CCL3, MIP-1β/CCL4, and RANTES/CCL5. These ex vivo results show, in a physiologically relevant model, that adenosine deaminase is able to enhance HIV-1 effector responses markedly. The possibility to revert regulatory T cell-mediated inhibition of immune responses by use of adenosine deaminase, an enzyme that hydrolyzes adenosine, merits attention for restoring T lymphocyte function in HIV-1 infection.
Collapse
Affiliation(s)
- Isaac Naval-Macabuhay
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Víctor Casanova
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Gemma Navarro
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Felipe García
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Agathe León
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Laia Miralles
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Cristina Rovira
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - José M Martinez-Navio
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Teresa Gallart
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Josefa Mallol
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - José M Gatell
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Carme Lluís
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Rafael Franco
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Peter J McCormick
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Núria Climent
- *Department of Biochemistry and Molecular Biology, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer-AIDS Research Group and Catalonian Center for HIV Vaccines, Barcelona, Spain; Infectious Diseases and AIDS Unit and Service of Immunology, Hospital Clínic de Barcelona, Spain; and School of Pharmacy, University of East Anglia, Norwich, Norfolk, United Kingdom
| |
Collapse
|
35
|
Shahbazi M, Zahedifard F, Taheri T, Taslimi Y, Jamshidi S, Shirian S, Mahdavi N, Hassankhani M, Daneshbod Y, Zarkesh-Esfahani SH, Papadopoulou B, Rafati S. Evaluation of Live Recombinant Nonpathogenic Leishmania tarentolae Expressing Cysteine Proteinase and A2 Genes as a Candidate Vaccine against Experimental Canine Visceral Leishmaniasis. PLoS One 2015. [PMID: 26197085 PMCID: PMC4509652 DOI: 10.1371/journal.pone.0132794] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Canine Visceral Leishmaniasis (CVL) is a major veterinary and public health problem caused by Leishmania infantum (L. infantum) in many endemic countries. It is a severe chronic disease with generalized parasite spread to the reticuloendothelial system, such as spleen, liver and bone marrow and is often fatal when left untreated. Control of VL in dogs would dramatically decrease infection pressure of L. infantum for humans, since dogs are the main domestic reservoir. In the past decade, various subunits and DNA antigens have been identified as potential vaccine candidates in experimental animal models, but none has been approved for human use so far. In this study, we vaccinated outbreed dogs with a prime-boost regimen based on recombinant L. tarentolae expressing the L. donovani A2 antigen along with cysteine proteinase genes (CPA and CPB without its unusual C-terminal extension (CPB-CTE) and evaluated its immunogenicity and protective immunity against L. infantum infectious challenge. We showed that vaccinated animals produced significantly higher levels of IgG2, but not IgG1, and also IFN-γ and TNF-α, but low IL-10 levels, before and after challenge as compared to control animals. Protection in dogs was also correlated with a strong DTH response and low parasite burden in the vaccinated group. Altogether, immunization with recombinant L. tarentolae A2-CPA-CPB-CTE was proven to be immunogenic and induced partial protection in dogs, hence representing a promising live vaccine candidate against CVL.
Collapse
MESH Headings
- Animals
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Cells, Cultured
- Cysteine Proteases/genetics
- Cysteine Proteases/immunology
- Dog Diseases/immunology
- Dog Diseases/parasitology
- Dog Diseases/prevention & control
- Dogs
- Female
- Gene Expression
- Immunity, Humoral
- Leishmania/enzymology
- Leishmania/genetics
- Leishmania/immunology
- Leishmaniasis Vaccines/immunology
- Leishmaniasis Vaccines/isolation & purification
- Leishmaniasis Vaccines/therapeutic use
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/prevention & control
- Leishmaniasis, Visceral/veterinary
- Male
- Vaccination/methods
- Vaccination/veterinary
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Attenuated/therapeutic use
Collapse
Affiliation(s)
- Mehdi Shahbazi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, 69 Pasteur Ave., Tehran, 13164, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farnaz Zahedifard
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, 69 Pasteur Ave., Tehran, 13164, Iran
| | - Tahereh Taheri
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, 69 Pasteur Ave., Tehran, 13164, Iran
| | - Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, 69 Pasteur Ave., Tehran, 13164, Iran
| | - Shahram Jamshidi
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Sadegh Shirian
- Department of Molecular and Cytopathology, Daneshbod Pathology Laboratory, Shiraz, Iran
| | - Niousha Mahdavi
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Hassankhani
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Yahya Daneshbod
- Department of Molecular and Cytopathology, Daneshbod Pathology Laboratory, Shiraz, Iran
| | | | - Barbara Papadopoulou
- Research Center in Infectious Diseases, CHU de Québec Research Center and Department of Microbiology, Infectious Disease and Immunology, Laval University, Quebec, QC, Canada
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, 69 Pasteur Ave., Tehran, 13164, Iran
- * E-mail:
| |
Collapse
|
36
|
Sánchez-Valdéz FJ, Pérez Brandán C, Ferreira A, Basombrío MÁ. Gene-deleted live-attenuated Trypanosoma cruzi parasites as vaccines to protect against Chagas disease. Expert Rev Vaccines 2014; 14:681-97. [PMID: 25496192 DOI: 10.1586/14760584.2015.989989] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi. This illness is now becoming global, mainly due to congenital transmission, and so far, there are no prophylactic or therapeutic vaccines available to either prevent or treat Chagas disease. Therefore, different approaches aimed at identifying new protective immunogens are urgently needed. Live vaccines are likely to be more efficient in inducing protection, but safety issues linked with their use have been raised. The development of improved protozoan genetic manipulation tools and genomic and biological information has helped to increase the safety of live vaccines. These advances have generated a renewed interest in the use of genetically attenuated parasites as vaccines against Chagas disease. This review discusses the protective capacity of genetically attenuated parasite vaccines and the challenges and perspectives for the development of an effective whole-parasite Chagas disease vaccine.
Collapse
|
37
|
Joshi S, Rawat K, Yadav NK, Kumar V, Siddiqi MI, Dube A. Visceral Leishmaniasis: Advancements in Vaccine Development via Classical and Molecular Approaches. Front Immunol 2014; 5:380. [PMID: 25202307 PMCID: PMC4141159 DOI: 10.3389/fimmu.2014.00380] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 07/24/2014] [Indexed: 11/13/2022] Open
Abstract
Visceral leishmaniasis (VL) or kala-azar, a vector-borne protozoan disease, shows endemicity in larger areas of the tropical, subtropical and the Mediterranean countries. WHO report suggested that an annual incidence of VL is nearly 200,000 to 400,000 cases, resulting in 20,000 to 30,000 deaths per year. Treatment with available anti-leishmanial drugs are not cost effective, with varied efficacies and higher relapse rate, which poses a major challenge to current kala-azar control program in Indian subcontinent. Therefore, a vaccine against VL is imperative and knowing the fact that recovered individuals developed lifelong immunity against re-infection, it is feasible. Vaccine development program, though time taking, has recently gained momentum with the emergence of omic era, i.e., from genomics to immunomics. Classical as well as molecular methodologies have been overtaken with alternative strategies wherein proteomics based knowledge combined with computational techniques (immunoinformatics) speed up the identification and detailed characterization of new antigens for potential vaccine candidates. This may eventually help in the designing of polyvalent synthetic and recombinant chimeric vaccines as an effective intervention measures to control the disease in endemic areas. This review focuses on such newer approaches being utilized for vaccine development against VL.
Collapse
Affiliation(s)
- Sumit Joshi
- Division of Parasitology, Central Drug Research Institute , Lucknow , India
| | - Keerti Rawat
- Division of Parasitology, Central Drug Research Institute , Lucknow , India
| | | | - Vikash Kumar
- Division of Molecular and Structural Biology, Central Drug Research Institute , Lucknow , India
| | - Mohammad Imran Siddiqi
- Division of Molecular and Structural Biology, Central Drug Research Institute , Lucknow , India
| | - Anuradha Dube
- Division of Parasitology, Central Drug Research Institute , Lucknow , India
| |
Collapse
|
38
|
Saljoughian N, Taheri T, Rafati S. Live vaccination tactics: possible approaches for controlling visceral leishmaniasis. Front Immunol 2014; 5:134. [PMID: 24744757 PMCID: PMC3978289 DOI: 10.3389/fimmu.2014.00134] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 03/17/2014] [Indexed: 01/15/2023] Open
Abstract
Vaccination with durable immunity is the main goal and fundamental to control leishmaniasis. To stimulate the immune response, small numbers of parasites are necessary to be presented in the mammalian host. Similar to natural course of infection, strategy using live vaccine is more attractive when compared to other approaches. Live vaccines present the whole spectrum of antigens to the host immune system in the absence of any adjuvant. Leishmanization was the first effort for live vaccination and currently used in a few countries against cutaneous leishmaniasis, in spite of their obstacle and safety. Then, live attenuated vaccines developed with similar promotion of creating long-term immunity in the host with lower side effect. Different examples of attenuated strains are generated through long-term in vitro culturing, culturing under drug pressure, temperature sensitivity, and chemical mutagenesis, but none is safe enough and their revision to virulent form is possible. Attenuation through genetic manipulation and disruption of virulence factors or essential enzymes for intracellular survival are among other approaches that are intensively under study. Other designs to develop live vaccines for visceral form of leishmaniasis are utilization of live avirulent microorganisms such as Lactococcus lactis, Salmonella enterica, and Leishmania tarentolae called as vectored vaccine. Apparently, these vaccines are intrinsically safer and can harbor the candidate antigens in their genome through different genetic manipulation and create more potential to control Leishmania parasite as an intracellular pathogen.
Collapse
Affiliation(s)
- Noushin Saljoughian
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Tahareh Taheri
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Rafati
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
39
|
Opata MM, Stephens R. Early Decision: Effector and Effector Memory T Cell Differentiation in Chronic Infection. ACTA ACUST UNITED AC 2014; 9:190-206. [PMID: 24790593 PMCID: PMC4000274 DOI: 10.2174/1573395509666131126231209] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/08/2013] [Accepted: 11/19/2013] [Indexed: 11/22/2022]
Abstract
As effector memory T cells (Tem) are the predominant population elicited by chronic parasitic infections,
increasing our knowledge of their function, survival and derivation, as phenotypically and functionally distinct from
central memory and effector T cells will be critical to vaccine development for these diseases. In some infections, memory
T cells maintain increased effector functions, however; this may require the presence of continued antigen, which can also
lead to T cell exhaustion. Alternatively, in the absence of antigen, only the increase in the number of memory cells
remains, without enhanced functionality as central memory. In order to understand the requirement for antigen and the
potential for longevity or protection, the derivation of each type of memory must be understood. A thorough review of the
data establishes the existence of both memory (Tmem) precursors and effector T cells (Teff) from the first hours of an
immune response. This suggests a new paradigm of Tmem differentiation distinct from the proposition that Tmem only
appear after the contraction of Teff. Several signals have been shown to be important in the generation of memory T cells,
such as the integrated strength of “signals 1-3” of antigen presentation (antigen receptor, co-stimulation, cytokines) as
perceived by each T cell clone. Given that these signals integrated at antigen presentation cells have been shown to
determine the outcome of Teff and Tmem phenotypes and numbers, this decision must be made at a very early stage. It
would appear that the overwhelming expansion of effector T cells and the inability to phenotypically distinguish memory
T cells at early time points has masked this important decision point. This does not rule out an effect of repeated
stimulation or chronic inflammatory milieu on populations generated in these early stages. Recent studies suggest that
Tmem are derived from early Teff, and we suggest that this includes Tem as well as Tcm. Therefore, we propose a
testable model for the pathway of differentiation from naïve to memory that suggests that Tem are not fully differentiated
effector cells, but derived from central memory T cells as originally suggested by Sallusto et al. in 1999, but much
debated since.
Collapse
Affiliation(s)
- Michael M Opata
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Disease, 300 University Avenue, Galveston, TX 77555-0435, USA
| | - Robin Stephens
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Disease, 300 University Avenue, Galveston, TX 77555-0435, USA
| |
Collapse
|
40
|
Humanized HLA-DR4 mice fed with the protozoan pathogen of oysters Perkinsus marinus (Dermo) do not develop noticeable pathology but elicit systemic immunity. PLoS One 2014; 9:e87435. [PMID: 24498105 PMCID: PMC3909113 DOI: 10.1371/journal.pone.0087435] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 12/23/2013] [Indexed: 12/02/2022] Open
Abstract
Perkinsus marinus (Phylum Perkinsozoa) is a marine protozoan parasite responsible for “Dermo” disease in oysters, which has caused extensive damage to the shellfish industry and estuarine environment. The infection prevalence has been estimated in some areas to be as high as 100%, often causing death of infected oysters within 1–2 years post-infection. Human consumption of the parasites via infected oysters is thus likely to occur, but to our knowledge the effect of oral consumption of P. marinus has not been investigated in humans or other mammals. To address the question we used humanized mice expressing HLA-DR4 molecules and lacking expression of mouse MHC-class II molecules (DR4.EA0) in such a way that CD4 T cell responses are solely restricted by the human HLA-DR4 molecule. The DR4.EA0 mice did not develop diarrhea or any detectable pathology in the gastrointestinal tract or lungs following single or repeated feedings with live P. marinus parasites. Furthermore, lymphocyte populations in the gut associated lymphoid tissue and spleen were unaltered in the parasite-fed mice ruling out local or systemic inflammation. Notably, naïve DR4.EA0 mice had antibodies (IgM and IgG) reacting against P. marinus parasites whereas parasite specific T cell responses were undetectable. Feeding with P. marinus boosted the antibody responses and stimulated specific cellular (IFNγ) immunity to the oyster parasite. Our data indicate the ability of P. marinus parasites to induce systemic immunity in DR4.EA0 mice without causing noticeable pathology, and support rationale grounds for using genetically engineered P. marinus as a new oral vaccine platform to induce systemic immunity against infectious agents.
Collapse
|
41
|
Vintiñi EO, Medina M. Immune response in nasopharynx, lung, and blood elicited by experimental nasal pneumococcal vaccines containing live or heat-killed lactobacilli as mucosal adjuvants. Can J Physiol Pharmacol 2014; 92:124-31. [PMID: 24502635 DOI: 10.1139/cjpp-2013-0227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
This work analyzes the humoral and cellular immune responses induced by live (LcV) and heat-killed (LcM) Lactobacillus casei associated with the pneumococcal antigen (P-Ag) at the nasopharynx level, considering nasal-associated lymphoid tissue (NALT) as the primary inductive site of the mucosal immune system, and lung and blood as effector sites. Levels of P-Ag IgA and IgG antibodies, main types of B and T cells, and cytokines in mucosal and systemic compartments were evaluated. The results showed that both LcM+P-Ag and LcV+P-Ag vaccines effectively induced IgA and IgG anti-P-Ag Abs in the upper and lower respiratory tract and plasma. These results correlated with increased IgA+ cells in NALT and lung that was induced by the experimental vaccines. Moreover, numbers of IgG+ cells increased in the blood. Profiles of inflammatory and regulatory cytokines were evaluated and their possible implications for the defense against pneumococci was assessed. Considering the overall results, the potential mechanisms of immune stimulation induced by LcM and LcV used as adjuvants are discussed. LcV and LcM showed similar effects on the immune system. Strain viability is not crucial for the stimulation of the antigen-specific immune response, and LcM is a convenient and effective mucosal adjuvant.
Collapse
Affiliation(s)
- Elisa O Vintiñi
- a Facultad de Agronomía y Zootecnia, Florentino Ameghino S/N, El Manantial, Universidad Nacional de Tucumán, Tucumán, Argentina
| | | |
Collapse
|
42
|
Haddad R, Saldanha-Araujo F. Mechanisms of T-cell immunosuppression by mesenchymal stromal cells: what do we know so far? BIOMED RESEARCH INTERNATIONAL 2014; 2014:216806. [PMID: 25025040 PMCID: PMC4082893 DOI: 10.1155/2014/216806] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 05/15/2014] [Accepted: 05/31/2014] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells, which can give rise to several cell types including osteoblasts, adipocytes, and chondroblasts. These cells can be found in a variety of adult and fetal tissues, such as bone marrow, adipose tissue, cord blood, and placenta. In recent years, the biological properties of MSCs have attracted the attention of researchers worldwide due to their potential application for treating a series of clinical situations. Among these properties, special attention should be given to the immunoregulatory potential of those cells. MSCs are able to act on all cells of the immune system, which includes the capacity to inhibit the proliferation and function of T-cells. This feature renders them natural candidates to treat several diseases in which cellular immune response is exacerbated. In this review, we outline the main mechanisms by which MSCs immunosuppress T-cell response, focusing on cell-cell contact, secretion of soluble factors, and regulatory T-cell generation. The influence of surface markers in the immunosuppression process and features of MSCs isolated from different sources are also discussed. Finally, the influences of toll-like receptors and cytokines on the inflammatory microenvironment are highlighted regarding the activation of MSCs to exert their immunoregulatory function.
Collapse
Affiliation(s)
- Rodrigo Haddad
- 1Faculty of Ceilandia, University of Brasilia, 72220-900 Brasilia, DF, Brazil
| | - Felipe Saldanha-Araujo
- 2Faculty of Health Sciences, University of Brasilia, 70910-900 Brasilia, DF, Brazil
- *Felipe Saldanha-Araujo:
| |
Collapse
|
43
|
Liu Q, Wang F, Wang G, Zhao Q, Min J, Wang S, Cong H, Li Y, He S, Zhou H. Toxoplasma gondii: immune response and protective efficacy induced by ROP16/GRA7 multicomponent DNA vaccine with a genetic adjuvant B7-2. Hum Vaccin Immunother 2013; 10:184-91. [PMID: 24096573 DOI: 10.4161/hv.26703] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Toxoplasma gondii infection occurs commonly in humans and other warm-blooded animals. Its serious impact on public health and livestock sectors makes the development of an effective vaccine particularly important. In the current study, we constructed a multiantigenic DNA vaccine expressing ROP16 and GRA7 of T. gondii and evaluated the protective efficacy of these two fragments with or without a plasmid encoding murine costimulatory molecule B7-2. These recombinant eukaryotic expression plasmids were termed pROP16, pGRA7, pROP16-GRA7 and pB7-2, respectively. After intramuscular immunization in Kunming mice, we assessed the immune response using cytokine and antibody determinations, T lymphocyte subsets analysis, and the survival times of mice post acute T. gondii challenge. The results showed that mice immunized with the multiantigenic DNA vaccine pROP16-GRA7 gained higher levels of IgG titers and IgG2a subclass titers, production of IFN-γ, percentage of CD8+ T cells and median survival times against the acute infection of T. gondii compared with those of mice administered with pROP16 or pGRA7 and those in control groups. Moreover, the adjuvant pB7-2 formulated with DNA vaccine boosted these humoral and cellular (Th1, CD8+ T cell) immune responses. Therefore, it might be a promising genetic adjuvant to DNA vaccine against T. gondii for further investigation.
Collapse
Affiliation(s)
- Qi Liu
- Department of Parasitology; Shandong University School of Medicine; Jinan, Shandong PR China
| | - Fuwu Wang
- Department of Histology and Embryology; Shandong University School of Medicine; Jinan, Shandong PR China
| | - Guan Wang
- Department of Immunopharmacology and Immunotherapy; Shandong University School of Pharmaceutical Sciences; Jinan, Shandong PR China
| | - Qunli Zhao
- Department of Parasitology; Shandong University School of Medicine; Jinan, Shandong PR China
| | - Juan Min
- Wuhan Institute of Virology; Chinese Academy of Sciences; Wuhan, Hubei PR China
| | - Shuai Wang
- Department of Parasitology; Shandong University School of Medicine; Jinan, Shandong PR China
| | - Hua Cong
- Department of Parasitology; Shandong University School of Medicine; Jinan, Shandong PR China
| | - Ying Li
- Department of Parasitology; Shandong University School of Medicine; Jinan, Shandong PR China
| | - Shenyi He
- Department of Parasitology; Shandong University School of Medicine; Jinan, Shandong PR China
| | - Huaiyu Zhou
- Department of Parasitology; Shandong University School of Medicine; Jinan, Shandong PR China
| |
Collapse
|
44
|
Chen CY, Lin SY, Cheng MC, Tsai CP, Hung CL, Lo KW, Hwang Y, Hu YC. Baculovirus vector as an avian influenza vaccine: hemagglutinin expression and presentation augment the vaccine immunogenicity. J Biotechnol 2013; 164:143-50. [PMID: 23313887 DOI: 10.1016/j.jbiotec.2012.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 12/17/2012] [Accepted: 12/21/2012] [Indexed: 12/11/2022]
Abstract
Baculovirus simultaneously displaying and expressing the avian influenza virus (AIV) hemagglutinin (HA) protein can induce potent anti-HA humoral and cellular immune responses. Based on the hypothesis that improving the antigen expression and presentation can further boost the AIV vaccine efficacies, we first constructed a baculoviral vector (Bac-HAW) with HA gene fused with the woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) near its 3' end and expressed under the control of the hybrid CAG promoter. The WPRE fusion improved the HA expression and augmented the humoral and Th1 cellular immune responses after intramuscular administration into BALB/c mice. With Bac-HAW as the backbone, we next constructed Bac-HAMW which harbored the HA gene flanked with the signal sequence (MHCIss) and trafficking domain (MITD) of MHC class I molecule. In comparison with Bac-HAW, Bac-HAMW ameliorated the HA peptide presentation, significantly elevated the HA-specific humoral response (total IgG, IgG2a and hemagglutination inhibition titers) and favorably boosted the Th1 and IFN-γ(+)/CD8(+) T cell responses without extraneous adjuvants. These data collectively confirmed that enhancement of antigen expression and presentation by combining the WPRE and MHCIss/MITD fusion can potentiate the immunogenicity of the baculovirus-based vaccine, and implicates the potential of Bac-HAMW as an appealing AIV vaccine.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Waeckerle-Men Y, Bruffaerts N, Liang Y, Jurion F, Sander P, Kündig TM, Huygen K, Johansen P. Lymph node targeting of BCG vaccines amplifies CD4 and CD8 T-cell responses and protection against Mycobacterium tuberculosis. Vaccine 2012; 31:1057-64. [PMID: 23273509 DOI: 10.1016/j.vaccine.2012.12.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/07/2012] [Accepted: 12/12/2012] [Indexed: 02/01/2023]
Abstract
Vaccination with Mycobacterium bovis BCG provides limited protection against pulmonary tuberculosis and a risk of dissemination in immune-compromised vaccinees. For the development of new TB vaccines that stimulate strong T-cell responses a variety of strategies is being followed, especially recombinant BCG and attenuated M. tuberculosis. The objective of the current study was to test potential benefits of vaccination through direct lymph-node targeting of wildtype BCG; the recommended route of vaccination with BCG is intradermal. C57BL/6 mice were immunised with BCG by intradermal, subcutaneous or intralymphatic injections. Cellular immune responses and protection against M. tuberculosis were determined. Intralymphatic vaccination was 100-1000 times more effective in stimulating BCG-specific immune responses than intradermal or subcutaneous immunisation. Intralymphatic administration stimulated high frequencies of mycobacterium-specific lymphocytes with strong proliferating capacity and production of TNF-α, IL-2, IL-17 and, especially, IFN-γ secretion by. CD4 and CD8 T cells. Most importantly, intralymphatic vaccination with 2×10(3)CFU BCG induced sustained protection against M. tuberculosis in intratracheally challenged C57BL/6 mice, whereas subcutaneous vaccination with 2×10(5)CFU BCG conferred only a transient protection. Hence, direct administration of M. bovis BCG to lymph nodes demonstrates that efficient targeting to lymph nodes may help to overcome the efficacy problems of vaccination with BCG.
Collapse
Affiliation(s)
- Ying Waeckerle-Men
- Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Ignatowicz L, Mazurek J, Leepiyasakulchai C, Sköld M, Hinkula J, Källenius G, Pawlowski A. Mycobacterium tuberculosis infection interferes with HIV vaccination in mice. PLoS One 2012; 7:e41205. [PMID: 22848444 PMCID: PMC3406616 DOI: 10.1371/journal.pone.0041205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/18/2012] [Indexed: 01/28/2023] Open
Abstract
Tuberculosis (TB) has emerged as the most prominent bacterial disease found in human immunodeficiency virus (HIV)-positive individuals worldwide. Due to high prevalence of asymptomatic Mycobacterium tuberculosis (Mtb) infections, the future HIV vaccine in areas highly endemic for TB will often be administrated to individuals with an ongoing Mtb infection. The impact of concurrent Mtb infection on the immunogenicity of a HIV vaccine candidate, MultiHIV DNA/protein, was investigated in mice. We found that, depending on the vaccination route, mice infected with Mtb before the administration of the HIV vaccine showed impairment in both the magnitude and the quality of antibody and T cell responses to the vaccine components p24Gag and gp160Env. Mice infected with Mtb prior to intranasal HIV vaccination exhibited reduced p24Gag-specific serum IgG and IgA, and suppressed gp160Env-specific serum IgG as compared to respective titers in uninfected HIV-vaccinated controls. Importantly, in Mtb-infected mice that were HIV-vaccinated by the intramuscular route the virus neutralizing activity in serum was significantly decreased, relative to uninfected counterparts. In addition mice concurrently infected with Mtb had fewer p24Gag-specific IFN-γ-expressing T cells and multifunctional T cells in their spleens. These results suggest that Mtb infection might interfere with the outcome of prospective HIV vaccination in humans.
Collapse
Affiliation(s)
- Lech Ignatowicz
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jolanta Mazurek
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Markus Sköld
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jorma Hinkula
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Gunilla Källenius
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
| | - Andrzej Pawlowski
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
47
|
Taylor JL, Wieczorek A, Keyser AR, Grover A, Flinkstrom R, Karls RK, Bielefeldt-Ohmann H, Dobos KM, Izzo AA. HspX-mediated protection against tuberculosis depends on its chaperoning of a mycobacterial molecule. Immunol Cell Biol 2012; 90:945-54. [PMID: 22801575 PMCID: PMC3511932 DOI: 10.1038/icb.2012.34] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
New approaches consisting of ‘multistage' vaccines against (TB) are emerging that combine early antigenic proteins with latency-associated antigens. In this study, HspX was tested for its potential to elicit both short- and long-term protective immune responses. HspX is a logical component in vaccine strategies targeting protective immune responses against primary infection, as well as against reactivation of latent infection, because as previously shown, it is produced during latency, and as our studies show, it elicits protection within 30 days of infection. Recent studies have shown that the current TB vaccine, bacilli Calmette-Guerin (BCG), does not induce strong interferon-γ T-cell responses to latency-associated antigens like HspX, which may be in part why BCG fails to protect against reactivation disease. We therefore tested HspX protein alone as a prophylactic vaccine and as a boost to BCG vaccination, and found that HspX purified from M. tuberculosis cell lysates protected mice against aerosol challenge and improved the protective efficacy of BCG when used as a booster vaccine. Native HspX was highly immunogenic and protective, in a dose-dependent manner, in both short- and long-term infection models. Based on these promising findings, HspX was produced as a recombinant protein in E. coli, as this would enable facile purification; however, recombinant HspX (rHspX) alone consistently failed to protect against aerosol challenge. Incubation of rHspX with mycobacterial cell lysate and re-purification following incubation restored the capacity of the protein to confer protection. These data suggest the possibility that the native form may chaperone an immunogenic and protective antigen that is mycobacteria-specific.
Collapse
Affiliation(s)
- Jennifer L Taylor
- Department of Microbiology, Mycobacteria Research Laboratories, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523-1682, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Antigen-specific memory T cell responses after vaccination with an oral killed cholera vaccine in Bangladeshi children and comparison to responses in patients with naturally acquired cholera. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1304-11. [PMID: 22739692 DOI: 10.1128/cvi.00196-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Young children, older children, and adults develop comparable levels and durations of immunity following cholera. In comparison, young children receiving oral killed cholera vaccines (OCV) develop a lower level and shorter duration of protection than those of older children and adults. The reasons for this are unclear. We investigated OCV-induced memory T cell responses in younger and older children and compared responses to those in children with cholera. We found that patients with cholera developed significant levels of toxin-specific effector memory T cells (T(EM)) with follicular helper and gut-homing characteristics. Older children (6 to 14 years of age) receiving two doses of OCV containing recombinant cholera toxin B subunit (rCTB) had more modest T(EM) responses with follicular helper and gut-homing characteristics, but younger vaccinees (24 to 71 months of age) did not develop T(EM) responses. The T(EM) response correlated positively with subsequent IgG memory B cell responses specific to rCTB in older vaccinees. Cytokine analyses indicated that cholera patients developed significant Th1, Th17, and Th2 responses, while older children receiving vaccine developed more modest increases in Th1 and Th17 cells. Younger vaccinees had no increase in Th1 cells, a decrease in Th17 cells, and an increase in regulatory T (Treg) cells. Our findings suggest that T cell memory responses are markedly diminished in children receiving OCV, especially young children, compared to responses following naturally acquired cholera, and that these differences affect subsequent development of memory B cell responses. These findings may explain the lower efficacy and shorter duration of protection afforded by OCV in young children.
Collapse
|
49
|
DE ALMEIDA MARCOSC, MOREIRA HELMARN. A MATHEMATICAL MODEL OF IMMUNE RESPONSE IN CUTANEOUS LEISHMANIASIS. J BIOL SYST 2011. [DOI: 10.1142/s0218339007002209] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The TH1/TH2 paradigm has been largely used in the interpretation of several diseases, particularly in leishmaniasis. As far as we know there is no mathematical description of this model related to leishmaniasis. We have extended and modified a previous published set of equations1in order to adapt it to leishmanial disease particularities. The main modifications were: (1) the analysis of logistic and exponential parasite growth curves, (2) the assumption of the TH2 arm of the immune response having a positive action on parasite growth. The set of three simultaneous differential equations describing the TH1 arm, TH2 arm and parasite growth were analyzed for conditions of existence and stability of the solutions.Stable solutions valid for the logistic and exponential parasite growth models, with its possible clinical correlations, were obtained in the following situations: (1) parasite and TH2 extinction [TH1 cure], (2) parasite extinction and TH1/TH2 co-existence [TH1/TH2 cure], (3) TH1 and parasite co-existence, TH2 extinction [stable TH1 infection], and (4) TH1, TH2 and parasite co-existence [stable TH1/TH2 infection]. TH2 and parasite co-existence associated to TH1 extinction [stable TH2 infection] was obtained only with the logistic growth model. The model also provides an alternative hypothesis for TH1 bias in resistant mice and emphazises the importance of natural immunity for the existence of chronic states.
Collapse
Affiliation(s)
| | - HELMAR N. MOREIRA
- Department of Mathematics, Universidade de Brasilia, Brasilia-DF, CEP: 70910-900, Brazil
| |
Collapse
|
50
|
Simhadri VR, Mariano JL, Zhou Q, DeBell KE, Borrego F. Differential expression of CD300a/c on human TH1 and TH17 cells. BMC Immunol 2011; 12:62. [PMID: 22046970 PMCID: PMC3219710 DOI: 10.1186/1471-2172-12-62] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 11/02/2011] [Indexed: 11/29/2022] Open
Abstract
Background Human memory CD4+ T cells can be either CD300a/c+ or CD300a/c- and subsequent analyses showed that CD4+ effector memory T (TEM) cells are mostly CD300a/c+, whereas CD4+ central memory T (TCM) cells have similar frequencies of CD300a/c+ and CD300a/c- cells. Results Extensive phenotypical and functional characterization showed that in both TCM and TEM cells, the CD300a/c+ subset contained a higher number of TH1 (IFN-γ producing) cells. Alternatively, TH17 (IL-17a producing) cells tend to be CD300a/c-, especially in the TEM subset. Further characterization of the IL-17a+ cells showed that cells that produce only this cytokine are mostly CD300a/c-, while cells that produce IL-17a in combination with other cytokines, especially IFN-γ, are mostly CD300a/c+, indicating that the expression of this receptor is associated with cells that produce IFN-γ. Co-ligation of the TCR and CD300a/c in CD4+ T cells inhibited Ca2+ mobilization evoked by TCR ligation alone and modulated IFN-γ production on TH1 polarized cells. Conclusion We conclude that the CD300a/c receptors are differentially expressed on human TH1 and TH17 cells and that their ligation is capable of modulating TCR mediated signals.
Collapse
Affiliation(s)
- Venkateswara R Simhadri
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|